Hostname: page-component-788cddb947-r7bls Total loading time: 0 Render date: 2024-10-10T09:35:42.597Z Has data issue: false hasContentIssue false

Biological properties of vitamin B12

Published online by Cambridge University Press:  08 October 2024

Monika Moravcová
Affiliation:
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
Tomáš Siatka
Affiliation:
Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
Lenka Kujovská Krčmová
Affiliation:
Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic Department of Analytical Chemistry, Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
Kateřina Matoušová
Affiliation:
Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Hradec Králové, Czech Republic
Přemysl Mladěnka*
Affiliation:
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
*
*Corresponding author: Přemysl Mladěnka, email: mladenkap@faf.cuni.cz
Rights & Permissions [Opens in a new window]

Abstract

Vitamin B12, cobalamin, is indispensable for humans owing to its participation in two biochemical reactions: the conversion of l-methylmalonyl coenzyme A to succinyl coenzyme A, and the formation of methionine by methylation of homocysteine. Eukaryotes, encompassing plants, fungi, animals and humans, do not synthesise vitamin B12, in contrast to prokaryotes. Humans must consume it in their diet. The most important sources include meat, milk and dairy products, fish, shellfish and eggs. Due to this, vegetarians are at risk to develop a vitamin B12 deficiency and it is recommended that they consume fortified food. Vitamin B12 behaves differently to most vitamins of the B complex in several aspects, e.g. it is more stable, has a very specific mechanism of absorption and is stored in large amounts in the organism. This review summarises all its biological aspects (including its structure and natural sources as well as its stability in food, pharmacokinetics and physiological function) as well as causes, symptoms, diagnosis (with a summary of analytical methods for its measurement), prevention and treatment of its deficiency, and its pharmacological use and potential toxicity.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of The Nutrition Society

1. Introduction

Vitamins of the B complex are water-soluble molecules with essential roles in humans. Our previous paper comprehensively summarised the biological properties of vitamins B1, B2, B3 and B5 (Reference Hrubša, Siatka, Nejmanová, Vopršalová, Kujovská Krčmová and Matoušová1). Although there is a recent practical paper on vitamin B12 (Reference Wolffenbuttel, Owen, Ward and Green2), a comprehensive paper on vitamin B12 is missing.

Vitamin B12, cobalamin, with a molecular weight of 1355.4 Da, is indispensable for humans as an integral part of two biochemical reactions: the conversion of l-methylmalonyl coenzyme A to succinyl coenzyme A, and the formation of methionine by methylation of homocysteine. Humans, as well as animals and plants, are unable to synthesise it. Therefore, we have to obtain vitamin B12 from food(Reference Flippo and Holder3Reference Smith, Warren and Refsum6).

The compound was first identified as a nutrient or external factor in the 1920s thanks to the research efforts of Minot, Murphy and Whipple, who showed that the symptoms of pernicious anaemia can be overcome by adding liver to the diet. The structure of the compound was solved by Dorothy Hodgkin’s pioneering X-ray crystallography, which revealed that the vitamin was cyanolated, amidated, tetrapyrrole-containing cobalt. As a result, it was called cyanocobalamin(Reference Smith, Warren and Refsum6,Reference Martens, Barg, Warren and Jahn7) .

The structure of vitamin B12 is shown in Figure 1. The cobalt is located in the middle of a circular contraction of a modified tetrapyrrole macrocycle coordinated by four nitrogen atoms. This centre of the molecule is known as the corrin ring and is similar to, although quite different from, the tetrapyrrole-derived ring systems found in haem and chlorophylls. The lower nucleotide loop is bound to the corrin ring by a side chain attached to a macrocyclic ring that contains an unusual natural base 5,6-dimethylbenzimidazole, which also coordinates the cobalt ion. Thus, in cyanocobalamin, the cobalt ion is ligated not only by the four pyrrole nitrogens of the central ring, but also by the upper (β) and lower (α) ligands. The β-ligand in the molecule of B12 is a cyano group, while the α is nitrogen from the mentioned dimethylbenzimidazole. In biological systems, the upper cyano ligand is usually replaced by an adenosyl group to form adenosylcobalamin (more precisely, 5′-deoxyadenosylcobalamin, AdoCbl), or a methyl group to form methylcobalamin, or a hydroxyl group to form hydroxocobalamin. Similarly, some species use a different lower base where dimethylbenzimidazole is replaced by bases such as adenine, substituted benzimidazoles, which include hydroxy- or methoxybenzimidazole, or phenolic compounds, including phenol or cresol(Reference Smith, Warren and Refsum6,Reference Martens, Barg, Warren and Jahn7) .

Fig. 1. Structure of vitamin B12: Natural forms include 5′-deoxyadenosylcobalamin (AdoCbI), methylcobalamin and hydroxocobalamin, industrially produced is cyanocobalamin(Reference Martens, Barg, Warren and Jahn7). Structure was created by ChemDraw, version 20.0.

2. Sources of vitamin B12

Eukaryotes, encompassing plants, algae, protists, fungi, animals and humans, do not synthesise vitamin B12 (Reference Smith, Warren and Refsum6Reference Orłowska, Steczkiewicz and Muszewska22). Vitamin B12 is biosynthesised exclusively in prokaryotes. However, only about a third of all bacteria and archaea species are able to synthesise it(Reference Yilmaz and Walhout8,Reference Warren, Raux, Schubert and Escalante-Semerena9,Reference Kennedy and Taga23Reference Scott38) . These species provide it for other cobalamin non-producing bacteria and archaea(Reference Sokolovskaya, Shelton and Taga30,Reference Lauridsen, Matte and Lessard39Reference Doxey, Kurtz and Lynch51) as well as for eukaryotes, except plants and fungi which do not use vitamin B12 as an enzyme cofactor(Reference Smith, Warren and Refsum6,Reference Martens, Barg, Warren and Jahn7,Reference Sanudo-Wilhelmy, Gomez-Consarnau, Suffridge and Webb17,Reference Roth, Lawrence and Bobik18,Reference Sokolovskaya, Shelton and Taga30,Reference Osman, Cooke and Young52Reference Bertrand, Allen and Dupont58) .

Vitamin B12 is also produced by microbiota in the large intestine of humans, but it is not utilisable by the human body as it is not spatially bioavailable since the intrinsic factor-mediated absorption of cobalamin occurs in the upper part of the gastrointestinal tract (i.e. upstream of the location of its synthesis) as will be described in the following text related to pharmacokinetics(Reference Smith, Warren and Refsum6,Reference Sokolovskaya, Plessl and Bailey59Reference Biesalski69) . Moreover, a major proportion of the microbially produced vitamin B12 is utilised by other non-vitamin-producing microbes, further limiting its possible availability for the human host(Reference Degnan, Taga and Goodman60,Reference Biesalski69Reference Kundra, Geirnaert and Pugin76) . The latest observational study in adult humans reveals a slight colonic absorption of cobalamin (approximately 7%) and speculates about the potential involvement of the colonic microbiome in the body’s vitamin B12 homoeostasis(Reference Kurpad, Pasanna and Hegde77). However, the mechanism of vitamin B12 uptake from the colon and the overall contribution of the ‘endogenously produced’ colonic vitamin B12 to the maintenance of vitamin B12 status remain unknown(Reference Fedosov78). Based on the current knowledge, it seems that humans are mostly dependent on vitamin B12 from exogenous sources.

In animals, the vitamin B12 from prokaryotes is obtained and stored in tissues through microbial interactions in the natural food chain. A good example are ruminants, such as cattle and sheep. They are herbivores, i.e. they feed on pasture that does not contain cobalamin, but they obtain the essential vitamin B12 through symbiotic relationship with microbes inside their body. They have a specialised digestive organ positioned upstream of the small intestine – a rumen that is heavily colonised with various micro-organisms, including cobalamin-synthesising ones, and that allows fermentation of the ingested feed. The synthesised cobalamin is later absorbed in the small intestine, incorporated into organs and muscles, and secreted into milk(Reference Jiang, Lin, Xie, Jin, Zhu and Wang49,Reference Osman, Cooke and Young52,Reference Degnan, Taga and Goodman60,Reference Rowley and Kendall73,Reference Watanabe, Bito and Koseki79Reference Sobczyńska-Malefora, Delvin and McCaddon81) . Monogastric, non-ruminant herbivores, e.g. rabbits, receive the benefit of microbial cobalamin production in their own large intestine by consuming their faeces. This coprophagy enables microbial cobalamin absorption in the upper part of the digestive tract(Reference Osman, Cooke and Young52,Reference Degnan, Taga and Goodman60,Reference Rowley and Kendall73,Reference Antony82) . Omnivores, such as pigs and poultry, as well as carnivores acquire cobalamin from feed of animal origin(Reference Watanabe and Bito80,Reference Sobczyńska-Malefora, Delvin and McCaddon81) . In aquatic environments, cobalamin is produced only by certain bacteria and archaea. It is taken up by cobalamin-requiring bacteria as well as eukaryotic plankton, transferred to fish bodies via plankton, and concentrated in the larger piscivorous fishes. Similarly, the vitamin concentrates in the bodies of shellfish feeding on plankton(Reference Watanabe and Bito80,Reference Bito, Tanioka and Watanabe83Reference Watanabe, Yabuta, Tanioka and Bito85) .

Accordingly, foods of animal origin are the principal sources of vitamin B12 for humans(63,Reference Watanabe and Bito80,Reference González-Montaña, Escalera-Valente and Alonso86,Reference Franco-Lopez, Duplessis and Bui87) . The most important ones include meat, milk and dairy products, fish, shellfish and eggs(Reference Basu and Donaldson64,Reference Gille and Schmid68,Reference Bito, Tanioka and Watanabe83,Reference Watanabe84,88Reference Planells, Sanchez and Montellano136) . The highest levels of cobalamin are found in offal, especially the liver and kidney(Reference Basu and Donaldson64,Reference Gille and Schmid68,Reference Janice Marie Sych and Stevens70,Reference Allen, Miller and de Groot93,Reference Li111,Reference Truswell137Reference Azzini, Raguzzini and Polito141) . Shellfish, such as mussels, oysters, clams and shrimps, are rich in cobalamin. However, certain types of edible shellfish (herbivorous sea snails such as abalone and turban shell) are not suitable as a source of vitamin B12 because they contain substantial amounts of pseudocobalamin(Reference Watanabe and Bito80,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Watanabe and Bito142Reference Tanioka, Takenaka and Furusho147) . Pseudocobalamin (or pseudovitamin B12) is an analogue of cobalamin, in which adenine substitutes for 5,6-dimethylbenzimidazole as the lower axial ligand. It is biologically very likely inactive in humans because the intrinsic factor is very specific for binding cobalamin. Prevention of the absorption of cobalamin analogues might protect humans from their potential deleterious effects(Reference Smith, Warren and Refsum6,Reference Allen and Stabler74,Reference Watanabe, Bito and Koseki79,Reference Watanabe and Bito142,Reference Stupperich and NEXØ148Reference Stabler, Bernadette, Marriott and Yates152) . Interestingly, edible insects, such as mealworms, grasshoppers and cockroaches, might also be a source of vitamin B12 for humans. Crickets, however, do not suit this purpose because they contain pseudocobalamin as the predominant corrinoid compound(Reference Schmidt, Call, Macheiner and Mayer153Reference Bawa, Songsermpong, Kaewtapee and Chanput156).

Fungi, as mentioned above, neither produce nor utilise vitamin B12. Therefore, mushrooms and yeasts, e.g. black morels (Morchella conica), oyster mushrooms (Pleurotus ostreatus), parasol mushrooms (Macrolepiota procera), porcini mushrooms (Boletus sp.) and yeasts (Yarrowia lipolytica), generally contain none or very low amounts of this vitamin, which is presumably derived from the substrate on which they grow or from cobalamin-synthesising bacteria that live on the mushroom surface. Among edible mushrooms, the highest vitamin B12 contents were found in truffles (Tuber sp.), black trumpet (Craterellus cornucopioides), golden chanterelle (Cantharellus cibarius) and shiitake (Lentinula edodes)(Reference Bito, Teng and Ohishi14,Reference Watanabe and Bito80,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Watanabe, Schwarz and Takenaka157Reference Jach and Malm167) .

Algae, like other eukaryotic organisms, are not capable of synthesising vitamin B12 de novo. Over one half of all algal species require vitamin B12. Those algae, as well as cobalamin-independent ones, which can, however, also accumulate exogenous cobalamin, acquire the vitamin from prokaryotic producers that are in symbiotic relationships with algae or reside on algal surfaces(Reference Croft, Lawrence and Raux-Deery11,Reference Tang, Koch and Gobler12,Reference Ramanan, Kim and Cho53,Reference Kazamia, Czesnick and Nguyen56,Reference Watanabe and Bito80,Reference Bito, Tanioka and Watanabe83,Reference de Brito, Campos and Menezes158,Reference Brawley, Blouin and Ficko-Blean168Reference Kazamia, Helliwell, Purton and Smith179) . Changes in the character and magnitude of the epiphytic prokaryotic communities related to the region or algal physiological state (e.g. growing conditions and harvesting period) may contribute to variation in the vitamin content, but these factors are currently poorly quantified(Reference Wells, Potin and Craigie180). Contents of vitamin B12 vary interspecifically and intraspecifically in edible algae ranging from traces, e.g. in Irish moss (Chondrus crispus) and hijiki (Sargassum fusiforme), to substantial amounts, e.g. in green laver (Ulva spp.), purple laver or nori (Porphyra/Pyropia spp.) and Chlorella spp., especially the Chlorella grown non-aseptically under open culture conditions(Reference Watanabe and Bito80,Reference Bito, Tanioka and Watanabe83,Reference Kwak, Park and Cho144,Reference Bito, Teng and Watanabe150,Reference de Brito, Campos and Menezes158,Reference Bito, Bito and Asai172,Reference Kanazawa176,Reference Watanabe, Takenaka and Katsura181Reference Cherry, O’Hara and Magee193) .

Plants, like fungi, neither synthesise nor use vitamin B12 in their metabolism(Reference Roth, Lawrence and Bobik18,Reference Smith, Croft, Moulin and Webb20) . A few exceptions of plants containing some cobalamin have been reported: edible duckweed Wolffia globosa called mankai(Reference Kaplan, Zelicha and Tsaban194,Reference Sela, Yaskolka Meir and Brandis195) , sea buckthorn (Hippophae rhamnoides), elecampane (Inula helenium), couch grass (Elymus repens)(Reference Nakos, Pepelanova and Beutel21,Reference Kysil61) and tea plant (Camellia sinensis)(Reference Kittaka-Katsura, Watanabe and Nakano196). Cobalamin in those plants is, like in other eukaryotes, of prokaryotic origin and is presumably produced by symbiotic endophytic microbes(Reference Nakos, Pepelanova and Beutel21,Reference Kaplan, Zelicha and Tsaban194,Reference Sela, Yaskolka Meir and Brandis195) or taken up from soil containing some organic fertilisers such as fishmeal or manure(Reference Antony82,Reference Kittaka-Katsura, Watanabe and Nakano196,Reference Lawrence, Nemoto-Smith and Deery197) . Therefore, cobalamin is not a normal constituent of commonly eaten plant foods unless they are contaminated with cobalamin-producing microbes (e.g. from soil or manure), contain yeasts or have been exposed to microbial fermentation that have produced the vitamin, or have been fortified with cobalamin (e.g. fortified ready-to-eat breakfast cereals and bread)(Reference Smith, Warren and Refsum6,Reference Nakos, Pepelanova and Beutel21,Reference Sobczyńska-Malefora, Delvin and McCaddon81,Reference Antony82,Reference Watanabe84,88Reference Tucker, Rich and Rosenberg90,Reference Lavriša, Hristov and Hribar104,Reference Truswell137,Reference Stabler, Bernadette, Marriott and Yates152,Reference Ofoedu, Iwouno and Ofoedu198Reference Yajnik, Deshpande and Lubree202) .

Thus, individuals who consume diets completely free of animal products (vegans) and even lacto-ovo vegetarians are at risk of vitamin B12 deficiency compared with omnivores. Indeed, different national nutritional societies quote a need to ensure a reliable source of vitamin B12 in persons on plant-based diets(63,Reference Allen66,Reference Sobczyńska-Malefora, Delvin and McCaddon81,Reference Ströhle, Richter and González-Gross102,Reference Lavriša, Hristov and Hribar104,Reference Obeid, Heil and Verhoeven108,Reference Rizzo, Laganà and Rapisarda163,Reference Allen203Reference Niklewicz, Smith and Smith244) . Some dietary ingredients or food supplements of non-animal origin may be useful for vegetarians to partly contribute to the supply of vitamin B12. Microalgae Chlorella and Spirulina (Arthrospira) are the commercially most produced microalgal genera as dietary supplements (Reference Pulz and Gross245,Reference Lafarga, Fernández-Sevilla, González-López and Acién-Fernández246) . In particular, the green alga Chlorella containing cobalamin is a relevant source of it. On the contrary, microalgae, e.g. Spirulina, Aphanizomenon and Nostoc, i.e. cyanobacteria, contain predominantly pseudocobalamin that they synthesise, and only minor amounts of cobalamin acquired from the environment; therefore, they are not suitable sources of vitamin B12. It should also be emphasised that nutrient labels on products often do not differentiate between forms of vitamin B12 – i.e. they do not specify that pseudocobalamin is present there instead of cobalamin(Reference Helliwell, Lawrence and Holzer13,Reference Kennedy and Taga23,Reference Heal, Qin and Ribalet24,Reference Watanabe84,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Ströhle, Richter and González-Gross102,Reference Watanabe and Bito142,Reference Rizzo, Laganà and Rapisarda163,Reference van den Oever and Mayer171,Reference Bito, Bito and Asai172,Reference Wells, Potin and Craigie180,Reference Watanabe, Takenaka and Kittaka-Katsura182,Reference Watanabe, Yabuta, Bito and Teng184,Reference Kittaka-Katsura, Fujita, Watanabe and Nakano189,Reference Edelmann, Aalto and Chamlagain191,Reference Merchant, Phillips and Udani192,Reference Watanabe, Katsura and Takenaka247Reference Castillejo, Martínez-Hernández and Goffi253) . Macroalgae (seaweeds) green laver and purple laver, the most widely consumed edible algae, belong to the best non-animal sources of cobalamin; consumption of approximately 4 g of dried purple laver could supply the US recommended dietary allowance of 2.4 μg/d(Reference Watanabe and Bito80,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Kwak, Lee and Lee115,Reference Bito, Teng and Watanabe150,Reference de Brito, Campos and Menezes158,Reference Watanabe, Takenaka and Katsura181,Reference Watanabe, Takenaka and Kittaka-Katsura182,Reference Watanabe, Yabuta, Bito and Teng184,Reference Bito and Watanabe185,Reference Miyamoto, Yabuta and Kwak187,Reference Martínez–Hernández, Castillejo and Carrión–Monteagudo188,Reference Cherry, O’Hara and Magee193,Reference Cornish, Critchley and Mouritsen254Reference Green, Allen and Bjørke-Monsen260) . Shiitake can serve as a source of cobalamin. Nevertheless, although about 50 g of dried fruiting bodies could be adequate to achieve the daily cobalamin requirement, ingestion of such large amounts would not be feasible daily(Reference Bito, Teng and Ohishi14,Reference Watanabe and Bito80,Reference de Brito, Campos and Menezes158,Reference Marczykowski and Breidenassel259) . Sea buckthorn berries as well as mankai contain acceptable quantities of cobalamin; approximately 18 g of sea buckthorn jam (about 1.5 tablespoons), 6.5 g of sea buckthorn dried berries or circa 100 g of dried mankai (equivalent to 500 g of frozen mankai for making five cups of green shakes) may cover the daily US recommended amount of the vitamin specified above(Reference Nakos, Pepelanova and Beutel21,Reference Sela, Yaskolka Meir and Brandis195,Reference Jedut, Szwajgier, Glibowski and Iłowiecka261) . However, the cobalamin contents in all these alternative sources fluctuate greatly(Reference Bito, Teng and Ohishi14,Reference Nakos, Pepelanova and Beutel21,Reference Kysil61,Reference Watanabe and Bito80,Reference de Brito, Campos and Menezes158,Reference Teng, Bito and Takenaka160,Reference Rizzo, Laganà and Rapisarda163,Reference Bito, Bito and Asai172,Reference Kittaka-Katsura, Fujita, Watanabe and Nakano189) . Moreover, safety hazards posed by some constituents of those dietary products should not be underestimated. Continuous intake of large amounts of those products might adversely affect human health owing to the presence of potentially harmful substances, whose content should be monitored during the quality control(Reference Sánchez-Parra, Boutarfa and Aboal262Reference Van Hassel, Ahn and Huybrechts267). High consumption of algae is associated with higher risks of deleterious effects due to excessive intake of some minerals that algae naturally accumulate, e.g. iodine, cancerogenic arsenic and toxic heavy metals(Reference Bito, Teng and Watanabe150,Reference Wells, Potin and Craigie180,Reference Cherry, O’Hara and Magee193,Reference Rogerson218,Reference Kristensen, Madsen and Hansen228,Reference Sandgruber, Gielsdorf and Baur249,Reference Sánchez-Parra, Boutarfa and Aboal262,Reference Rzymski, Niedzielski and Kaczmarek263,Reference Grosshagauer, Kraemer and Somoza265,Reference Muñoz, Díaz and Nelson268Reference Lähteenmäki-Uutela, Rahikainen and Camarena-Gómez284) . In addition, these products might be detrimental to human health because they may be contaminated with toxic cyanotoxins produced by some cyanobacteria(Reference Sánchez-Parra, Boutarfa and Aboal262,Reference Rzymski and Jaśkiewicz264Reference Van Hassel, Ahn and Huybrechts267,Reference Mendes, Navalho and Ferreira270,Reference Marles, Barrett and Barnes285) and with cancerogenic polycyclic aromatic hydrocarbons from the environment(Reference Grosshagauer, Kraemer and Somoza265,Reference Ampofo and Abbey266,Reference Muys, Sui and Schwaiger280) . Similarly to algae, excessive ingestion of mushrooms may threaten human health as a consequence of exceeding dietary exposure limits of heavy metals, arsenic and radionuclides(Reference Li, Wang and Wang286Reference Guillén and Baeza299). Consumption of Wolffia globosa (mankai) is of safety concern because it leads to an increase in manganese intake which could represent a risk of adverse health effects(300). Fermented plant-based foods, such as kimchi, sauerkraut, injera, kombucha, tempeh and miso, are generally poor dietary sources of vitamin B12 for vegetarians mainly because there is simply not enough vitamin B12 produced by these fermentations(Reference Nakos, Pepelanova and Beutel21,Reference Kennedy and Taga23,Reference Herbert65,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Kwak, Lee, Oh and Park114,Reference Kwak, Lee and Lee115,Reference de Brito, Campos and Menezes158,Reference Rizzo, Laganà and Rapisarda163,Reference Watanabe, Yabuta, Bito and Teng184,Reference Melina, Craig and Levin205,Reference Stabler and Allen214,Reference Kwak, Hwang, Watanabe and Park255,Reference Jedut, Szwajgier, Glibowski and Iłowiecka261,Reference Liem, Steinkraus and Cronk301Reference Masuda, Ide and Utsumi303) . Taking everything into account, all these alternative sources of cobalamin for vegetarians are unreliable, insufficient or impractical to meet cobalamin needs of the human body in the long term. They may improve vitamin B12 status in vegetarians but cannot replenish the total body store of the vitamin(Reference Watanabe, Yabuta, Tanioka and Bito85,Reference de Brito, Campos and Menezes158,Reference Watanabe, Yabuta, Bito and Teng184,Reference Merchant, Phillips and Udani192Reference Sela, Yaskolka Meir and Brandis195,Reference Melina, Craig and Levin205,Reference Zugravu, Macri, Belc and Bohiltea304309) .

Plant-based diets, except those rich in ultra-processed plant-based food products(Reference Gehring, Touvier and Baudry310Reference Orlich, Sabaté and Mashchak319), are considered potentially superior to a traditional omnivorous diet for reducing the risk of chronic diseases, such as metabolic syndrome with type 2 diabetes mellitus, hypertension, cardiovascular diseases in general, and several types of cancer(Reference Melina, Craig and Levin205,Reference Shaw, Zello and Rodgers217,Reference Phillips222,Reference Bakaloudi, Halloran and Rippin227,Reference Dawczynski, Weidauer and Richert241,Reference Craig, Mangels and Fresán320Reference Nhan, Sgambat and Moudgil344) . Meanwhile, compared with omnivorous diets, plant-based ones are deficient in some nutrients, which could have detrimental health implications as well. Vitamin B12 is of particular relevance(Reference Craig204,Reference Phillips222,Reference Bakaloudi, Halloran and Rippin227,Reference Schüpbach, Wegmüller and Berguerand231,Reference Dawczynski, Weidauer and Richert241Reference Niklewicz, Smith and Smith244,Reference Brown305,Reference Nebl, Schuchardt and Ströhle345,Reference Weikert, Trefflich and Menzel346) . The proposed benefit of plant-based-diets is conditioned by the balance and incorporation of all missing constituents so that nutritional quality is not compromised. Regular intake of cobalamin-fortified foods (mostly ready-to-eat-cereals, bread, nutritional yeast, meat analogues, such as tofu, and milk substitutes, e.g. soy, almond and rice milk) and/or cobalamin-containing supplements is recommended for people on plant-based diets to prevent vitamin B12 deficiency(Reference Janice Marie Sych and Stevens70,Reference Antony82,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Allen, Miller and de Groot93,Reference Azzini, Raguzzini and Polito141,Reference Rizzo, Laganà and Rapisarda163,Reference Watanabe, Yabuta, Bito and Teng184,Reference Melina, Craig and Levin205,Reference Rogerson218Reference Phillips222,Reference Zeuschner, Hokin and Marsh224Reference Woo, Kwok and Celermajer233,Reference White235,Reference Pawlak, Lester and Babatunde236,Reference Seves, Verkaik-Kloosterman, Biesbroek and Temme238,Reference Dawczynski, Weidauer and Richert241Reference Neufingerl and Eilander243,Reference Chandra-Hioe, Lee and Arcot256,Reference Marczykowski and Breidenassel259,Reference Brown305Reference Strain, Hughes, Pentieva, Biesalski, Drewnowski, Dwyer, Strain, Weber and Eggersdorfer308,Reference Craig, Mangels and Fresán320Reference Mangels, Messina and Melina326,Reference Nebl, Schuchardt and Ströhle345Reference Storz, Müller and Niederreiter373) . The vitamin B12 status should also be monitored regularly(Reference Ströhle, Richter and González-Gross102,Reference Azzini, Raguzzini and Polito141,Reference Rogerson218,Reference Herrmann, Schorr, Obeid and Geisel223,Reference Zeuschner, Hokin and Marsh224,Reference Bakaloudi, Halloran and Rippin227,Reference Woo, Kwok and Celermajer233,Reference White235,Reference Agnoli, Baroni and Bertini242,Reference Chandra-Hioe, Lee and Arcot256,Reference Richter, Boeing and Grünewald-Funk306,Reference Schwarz, Dschietzig and Schwarz307,Reference Craig, Mangels and Fresán320,Reference Jakše325,Reference Mangels, Messina and Melina326,Reference Donaldson347,Reference Rudloff, Bührer and Jochum360Reference Baroni, Goggi and Battaglino362,Reference Kiely367,Reference Hunt, Harrington and Robinson374) .

Contents of cobalamin in some selected foodstuffs are presented in Table 1.

Table 1. Cobalamin contents in selected foodstuffs

The stability of vitamin B12 in food products during processing, preparation and storage is an important parameter affecting the supply of the vitamin to consumers(Reference Janice Marie Sych and Stevens70,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Berry Ottaway, Skibsted, Risbo and Andersen375Reference Bennink and Ono378) . Cyanocobalamin is chemically more stable than adenosylcobalamin, methylcobalamin and hydroxocobalamin. In neutral and weakly acid aqueous solutions, it is stable at room temperature with highest stability at pH 4.5–5 and it is relatively stable to the thermal processing. Loss of vitamin activity may occur due to heat treatments under alkaline and strong acid conditions or due to light or via contact with reducing agents, such as ascorbic acid, polyphenols, nitrous oxide, sulphite and iron(II) salts, but also with oxidising ones, such as atmospheric oxygen, hypochlorous acid and chloramine-T(Reference Bito, Teng and Ohishi14,Reference Nakos, Pepelanova and Beutel21,Reference Gille and Schmid68,Reference Janice Marie Sych and Stevens70,Reference Li, Gill, Grainger and Manley-Harris124,Reference Watanabe and Bito142,Reference Teng, Bito and Takenaka160,Reference Ofoedu, Iwouno and Ofoedu198,Reference Berry Ottaway, Skibsted, Risbo and Andersen375Reference Bergström377,Reference Lee, Lee, Huh and Choi379Reference Temova Rakuša, Roškar, Hickey and Geremia393) . On the one hand, degradation of vitamin B12 caused by ascorbic acid might have little practical importance because foods containing vitamin B12 generally do not contain significant amounts of vitamin C(Reference Godoy, Amaya-Farfan, Rodriguez-Amaya, Rodriguez-Amaya and Amaya-Farfan376), but on the other hand, it should be taken into account, e.g. in vitamin fortified beverages(Reference Bajaj and Singhal388,Reference Yamada, Shimodaira and Chida390) .

In food matrices, vitamin B12 is generally considered to be rather heat stable compared with other water-soluble vitamins. Reported losses depend on the type of food and processing conditions(Reference Gille and Schmid68,Reference Janice Marie Sych and Stevens70,Reference Bodwell and Anderson134,Reference Bennink and Ono378,Reference Campo, Muela and Olleta394Reference Öhrvik, Carlsen, Källman and Martinsen406) . In milk, vitamin B12 is affected by heat processing; the more severe the process, the greater the loss is. Vitamin losses are generally less than 10% after pasteurisation, 10–20% after ultra-high temperature treatment, up to 20% following sterilisation and 20–35% during spray drying(Reference Watanabe84,Reference Berry Ottaway, Skibsted, Risbo and Andersen375,Reference Rolls and Porter396,Reference Oamen, Hansen and Swartzel407Reference Graham411) . The vitamin B12 amount decreased three times more in heated chocolate milk (by about 33%) than in the unflavoured one because cocoa powder used for milk flavouring contains polyphenols, which are mainly responsible for the decomposition of the vitamin(Reference Johns, Das and Kuil383). In meat, vitamin B12 is stable during the cooking, if the vitamin content in the cooking liquids, gravy and drippings is taken into consideration(Reference Berry Ottaway, Skibsted, Risbo and Andersen375,Reference Bognár403) . Indeed, most vitamin B12 losses result from leaching into water, e.g. during cooking or freezing/thawing(Reference Nishioka, Kanosue, Yabuta and Watanabe399,Reference Bognár403,Reference Lešková, Kubíková and Kováčiková412Reference Sato, Kudo and Muramatsu415) . This process is essentially similar to that of other water-soluble B vitamins(Reference Hrubša, Siatka, Nejmanová, Vopršalová, Kujovská Krčmová and Matoušová1). Boiling, stewing and frying lead to vitamin B12 losses of 20–40%, 10–40%, 30–50% and 10–20% in pork, beef, chicken and fish, respectively(Reference Bognár403). The highest reduction in the vitamin content occurs during boiling(Reference Bognár403Reference Öhrvik, Carlsen, Källman and Martinsen406). The best method for cooking fish was vacuum-packed pouch cooking with no loss of vitamin B12, compared with steaming, boiling, grilling, frying and microwaving(Reference Nishioka, Kanosue, Yabuta and Watanabe399). Scrambled, fried and hard-cooked eggs lose during cooking 5%, 5–15% and 20% vitamin B12, respectively(Reference Bognár403,Reference Öhrvik, Carlsen, Källman and Martinsen406) .

Treatment of foods with penetrating waves such as microwaves was shown to promote the degradation of vitamin B12 (Reference Janice Marie Sych and Stevens70,Reference Nishioka, Kanosue, Yabuta and Watanabe399,Reference Watanabe, Abe and Fujita416Reference Zheng, Xiang and Zhang418) . For instance, decreases by 17%, 14% and 48% were estimated in beef, pork and milk, respectively, treated with microwave heating for 6 min, which is a common time used for reheating of foods(Reference Watanabe, Abe and Fujita416).

Puffed rice extrudates could be used as a palatable vehicle for fortification with vitamin B12; losses of added vitamin ranged from 19% to 64% depending on extrusion processing parameters(Reference Bajaj and Singhal419). The stability of the added and in situ-produced vitamin B12 in breadmaking varied according to the chosen process (straight-, sponge- and sourdough processes)(Reference Edelmann, Chamlagain and Santin386,Reference Chamlagain420) . Converning baking from fortified whole wheat, the vitamin amounts were reduced by 9%, 20%, 66% and 76% in chapattis (unleavened flatbread), bread, cake and cookie, respectively. The lowest vitamin B12 retention was recorded in pooris prepared from fortified whole wheat flour by frying; the vitamin loss was 86%(Reference Bajaj and Singhal421).

Vitamin B12 is sensitive to light and ultraviolet (UV) radiation(Reference Janice Marie Sych and Stevens70,Reference Watanabe84,Reference Berry Ottaway, Skibsted, Risbo and Andersen375,Reference Godoy, Amaya-Farfan, Rodriguez-Amaya, Rodriguez-Amaya and Amaya-Farfan376,Reference Henry and Heppell381,Reference Yessaad, Bernard and Bourdeaux382,Reference Lie, Chandra-Hioe and Arcot384,Reference Juzeniene and Nizauskaite422,Reference Bito, Ohishi and Hatanaka423) . Sunlight at a brightness of 8000-foot candles (approximately 86 000 lux) caused a 10% loss of cyanocobalamin for each 30 min of exposure in neutral aqueous solutions, but exposures to levels of brightness below 300-foot candles (approximately 3200 lux) had little effect(Reference Berry Ottaway, Skibsted, Risbo and Andersen375). Photodegradation of cyanocobalamin in aqueous solutions may be accelerated by riboflavin, which acts as a sensitiser(Reference Godoy, Amaya-Farfan, Rodriguez-Amaya, Rodriguez-Amaya and Amaya-Farfan376,Reference Juzeniene and Nizauskaite422) . The vitamin B12 concentration in milk exposed to the light for 24 h decreased by 1–27%, depending on the type of milk tested(Reference Watanabe, Katsura, Abe and Nakano424), while no changes in the vitamin B12 content occurred in pasteurised milk packed in a clear polyethylene terephthalate bottle exposed to fluorescent light (1700 lux) for 10 d(Reference Saffert, Pieper and Jetten425). The photostability of vitamin B12 in foods may be increased due to matrix effects, such as binding to proteins(Reference Janice Marie Sych and Stevens70,Reference Zironi, Gazzotti and Barbarossa121,Reference Campos-Gimnez, Fontannaz and Trisconi426,Reference Wang, Shou and Zhu427) . Furthermore, light penetrates only slightly below the surface of foods, which would suggest that vitamin B12 photosensitivity is not a serious issue in most foods(Reference Janice Marie Sych and Stevens70).

Vitamin B12 content decreases in fermented milk products(Reference Scott and BISHOP397,Reference Arkbåge, Witthöft, Fondén and Jägerstad410) . Fermentation of milk resulted in vitamin B12 losses of 25% in yoghurt and 15% in Filmjölk. Storage of an unopened package of the final product at 4°C for 14 d, until the ‘use by date’, reduced the vitamin concentrations further by 33% and 26% for yoghurt and Filmjölk, respectively, so that they contained 40–60% of vitamin B12 originally present in the milk. This is most likely attributed to the consumption of the vitamin by starter cultures of lactic acid bacteria, which are metabolically active not only during fermentation but also at lower temperatures during storage(Reference Arkbåge, Witthöft, Fondén and Jägerstad410). During the cheese-making process, the whey fraction is removed, leading to a considerable loss of vitamin B12 (on average about 50% of the vitamin originally present in the milk) due to its water solubility. Meanwhile, the vitamin content in final products (especially in hard cheeses) is higher relative to the starting milk owing to the milk thickening during cheese production (for instance, about 10 litres of milk is required to produce 1 kg of hard cheese). Ripening and storage of cheeses do not alter the vitamin B12 content, except in mold cheeses, in which the content may decline(Reference Gille and Schmid68,Reference Arkbåge, Witthöft, Fondén and Jägerstad410,Reference Repossi, Zironi and Gazzotti428) . Swiss-type cheeses (e.g. Emmentaler and Gruyère) contain higher amounts of vitamin B12 than other ones owing to the application of propionibacteria as adjunct starter cultures for ripening (responsible for the characteristic flavor and opening formation) that are able to produce vitamin B12 (Reference Gille and Schmid68,Reference Matte, Britten and Girard99,Reference Arkbåge, Witthöft, Fondén and Jägerstad410,Reference Rabah, Carmo and Jan429,Reference Poonam and Tomar430) .

Maturation of meat between the time of slaughtering and consumption for up to 14 d does not affect the vitamin B12 content in beef(Reference Ortigues-Marty, Thomas and Prévéraud398).

Vitamin B12 is fairly stable to ionising radiation, which is used as a food preservation method to control foodborne pathogens and extend product shelf life; no losses of the vitamin were found in irradiated pork, chicken, clam and haddock(Reference Kilcast431Reference Fox, Thayer and Jenkins433). Hypochlorous acid water (used to sanitise food products, e.g. vegetables, fruits and meat) as well as sodium metabisulfite and sodium sulfite (used to prevent black discoloration of shrimps) destroy vitamin B12 in aqueous solutions but do not reduce its content in shrimps. Similarly, no significant changes in vitamin B12 amounts occur in beef treated with hypochlorous acid water. This is explained by the fact that most vitamin B12 present in foods is in protein-bound form rather than free(Reference Okamoto, Bito and Hiura385).

Some food ingredients have been shown to influence positively the stability of vitamin B12. Sorbitol, a sweetener, protects cyanocobalamin from degradation by heat, ascorbic acid, thiamine, UV light, and low or high pH values (Reference Lee, Lee, Huh and Choi379,Reference Lie, Chandra-Hioe and Arcot384) . Whey proteins enhance thermal stability of vitamin B12 by 20% and could be useful as protective agents against the physical destruction of the vitamin during food processing(Reference Wang, Shou and Zhu427). Carnosine, a dipeptide naturally present in meat, prevents the destruction of cyanocobalamin by vitamin C in the presence of copper ions and may be useful as an additive to multivitamin–mineral food supplements. Carnosine has been shown to possess antioxidant and metal chelating activity, which could be responsible for the observed protection(Reference Nakos, Pepelanova and Beutel21,Reference Takenaka, Sugiyama and Watanabe434) .

Storage may influence the rate of vitamin B12 decomposition(Reference Godoy, Amaya-Farfan, Rodriguez-Amaya, Rodriguez-Amaya and Amaya-Farfan376,Reference Bajaj and Singhal388,Reference Yamada, Shimodaira and Chida390,Reference Ford, Hurrell and Finot435,Reference Hemery, Fontan and Laillou436) . For instance, storage of ultra-high-temperature milk at room temperature for 18 weeks resulted in the complete disappearance of vitamin B12, probably due to exposure to dissolved oxygen in the container, while low temperature (7°C) did not alter the vitamin content for up to 18 weeks(Reference Gille and Schmid68,Reference Arkbåge131,Reference Godoy, Amaya-Farfan, Rodriguez-Amaya, Rodriguez-Amaya and Amaya-Farfan376,Reference Rolls and Porter396,Reference Oamen, Hansen and Swartzel407) . No appreciable losses of vitamin B12 were found in pasteurised milk during storage in a refrigerator for 9 d, regardless of how long it had been since the packages had been opened(Reference Andersson and Öste409). Effects of storage conditions (time, temperature, moisture content, oxygen and nitrogen) on the vitamin B12 amount in milk powders have also been studied(Reference Ford, Hurrell and Finot435). No remarkable changes in the vitamin B12 content occurred in vacuum-packaged salmon stored for 880 d at room temperature either on the Earth or exposed to spaceflight(Reference Zwart, Kloeris and Perchonok437). Cyanocobalamin loss reached up to 63% in fortified wheat flour packed in permeable paper bags, whereas no significant reduction of vitamin amounts occurred, when the flour was packed in multilayer aluminium/polyethylene bags (non-permeable to oxygen and humidity)(Reference Hemery, Fontan and Laillou436). When whole wheat flour fortified with cyanocobalamin was stored in air-tight plastic containers in the dark under different combinations of temperature (25°C and 45°C) and relative humidity (33%, 63% and 93%) to mimic the effects of various climatic conditions, the highest and lowest vitamin losses of 51% and 15% were recorded at 45°C/93% and at 25°C/all, respectively, after 120 d of storage, suggesting that wheat flour may be effectively fortified with vitamin B12 (Reference Bajaj and Singhal421). The degradation kinetics of vitamin B12 in fortified co-crystallised sugar cubes was studied under different storage conditions as for temperature and humidity; a half-life of 23 months was achieved at 25°C and 33% relative humidity(Reference Bajaj and Singhal438). Vitamin B12 was stable in a salt fortified with multiple micronutrients, including microencapsulated vitamins, during 6 months of storage(Reference Vinodkumar and Rajagopalan439). Different storage temperatures of fortified juices from carrot (pH 6) and lime (pH 2, richer in vitamin C promoting vitamin B12 degradation) showed losses of vitamin B12 of 8%, 15% and 19% in carrot juice and 82%, 95% and 100% in lime juice, respectively after 28 d at 4°C, 25°C and 37°C. Carrot juice is therefore more suitable for vitamin B12 fortification owing to its mild acidic character(Reference Bajaj and Singhal388).

Production of vitamin B12 and biofortification with B12

Vitamin B12, mainly in the most stable form cyanocobalamin, is commercially produced for use in fortified foods, dietary supplements, pharmaceuticals and animal feeds(Reference Hohmann, Litta and Hans67,Reference Sobczyńska-Malefora, Delvin and McCaddon81,88,Reference Allen, Miller and de Groot93,Reference Ströhle, Richter and González-Gross102,Reference Azzini, Raguzzini and Polito141,Reference Stabler, Bernadette, Marriott and Yates152,Reference Tucker, Olson and Bakun199,Reference Stabler and Allen214,Reference Chandra-Hioe, Lee and Arcot256,Reference Green, Allen and Bjørke-Monsen260,Reference Zugravu, Macri, Belc and Bohiltea304,Reference Damayanti, Jaceldo-Siegl and Beeson348,Reference Del Bo, Riso and Gardana351,Reference Lee, Lee, Huh and Choi379,Reference Obeid, Fedosov and Nexo440Reference de Benoist465) . The total synthesis of vitamin B12 comprising about seventy reactions was achieved in 1972(Reference Woodward466Reference Riether and Mulzer470). Due to the vitamin’s enormous structural complexity, its chemical synthesis is highly complicated and not economically feasible on an industrial scale. Therefore, industrial production of vitamin B12 is exclusively based on microbial fermentation. Currently, the most commonly employed micro-organisms are high-producing bacterial strains of Pseudomonas denitrificans and Propionibacterium freudenreichii, developed by means of random mutagenesis and selection, as well as genetic engineering from wild strains with high natural production ability(Reference Smith, Warren and Refsum6,Reference Martens, Barg, Warren and Jahn7,Reference Balabanova, Averianova, Marchenok, Son and Tekutyeva48,Reference Osman, Cooke and Young52,Reference Hohmann, Litta and Hans67,Reference Janice Marie Sych and Stevens70,Reference Pereira, Simões and Silva175,Reference Calvillo, Pellicer, Carnicer and Planas450,Reference Acevedo-Rocha, Gronenberg and Mack471Reference Kumar, Singh and Tiwari502) . Fermentations produce a mixture of hydroxocobalamin, adenosylcobalamin and methylcobalamin; they are then converted to cyanocobalamin by the addition of potassium cyanide(Reference Martens, Barg, Warren and Jahn7,Reference Balabanova, Averianova, Marchenok, Son and Tekutyeva48,Reference Janice Marie Sych and Stevens70,Reference Calvillo, Pellicer, Carnicer and Planas450,Reference Acevedo-Rocha, Gronenberg and Mack471,Reference Shimizu474,Reference Laudert, Hohmann and Moo-Young476,Reference Binod, Sindhu and Pandey478,Reference Survase, Bajaj and Singhal479) . Current industrial biotechnological processes for the production of vitamin B12 are suboptimal. Accordingly, further possible micro-organisms suitable for large-scale production have been widely studied in recent years, e.g. natural producers Bacillus megaterium and Sinorhizobium meliloti, and even Escherichia coli, which is not able to synthesise vitamin B12 de novo in nature but can do so after genetic modifications (heterologous expression of the whole biosynthetic pathway). However, the reported yields are not yet competitive with those achieved in present-day manufacturing bioprocesses(Reference Martens, Barg, Warren and Jahn7,Reference Balabanova, Averianova, Marchenok, Son and Tekutyeva48,Reference Calvillo, Pellicer, Carnicer and Planas450,Reference Acevedo-Rocha, Gronenberg and Mack471,Reference Wang, Liu, Jin and Zhang472,Reference Mani503Reference Xu, Xiao and Yu517) .

Biofortification aims to make crop plants naturally more nutritive rather than adding nutrient supplements to the foods during food processing. Biofortification can be achieved through three main approaches including plant breeding, transgenic techniques and agronomic practices(Reference Malik and Maqbool518Reference Oh, Cave and Lu522). No efforts are being made to biofortify crops with vitamin B12 through conventional breeding or genetic engineering due to the fact that the biosynthetic pathway is present exclusively in some bacteria and archaea(Reference Titcomb and Tanumihardjo521,Reference Garg, Sharma and Vats523) . As for the agronomic approach, which requires physical application of cobalamin to plants for enriching them with this vitamin, some biofortification attempts have been reported, e.g. in wheat and spinach(Reference Mozafar524) and green tea(Reference Kittaka-Katsura, Watanabe and Nakano196) by the addition of organic fertilisers (naturally rich in vitamin B12) to the soil, in garden cress seedlings by growing on an agar medium containing cobalamin(Reference Lawrence, Nemoto-Smith and Deery197), in Japanese radish sprout (kaiware daikon) by soaking its seeds in cobalamin solution(Reference Sato, Kudo and Muramatsu415), in lettuce(Reference Bito, Ohishi and Hatanaka423) and spinach(Reference Zheng, Xiang and Zhang418) in hydroponic culture by treatment with cobalamin, and in the recombinant alga Chlamydomonas (expressing human intrinsic factor) by culturing in a medium supplemented with cobalamin(Reference Lima, Webb and Deery525). Milk concentrations of vitamin B12 are influenced by the genotype of the cow. Genomic regions associated with vitamin B12 concentrations in milk have been identified, which offer an interesting potential for marker-assisted genetic selection and breeding to increase the content of vitamin B12 in cow milk. However, feeding composition management could help optimise vitamin B12 amounts in milk only to a limited extent as there are adequate cobalt levels in the cow’s diet required by ruminal microbiota for cobalamin biosynthesis(Reference González-Montaña, Escalera-Valente and Alonso86,Reference Duplessis, Pellerin, Cue and Girard97,Reference Matte, Britten and Girard99,Reference Rutten, Bouwman and Sprong526Reference Stemme, Lebzien, Flachowsky and Scholz531) . The introduction of vitamin B12 into foods may also be achieved via the in situ production by micro-organisms naturally capable of synthesising cobalamin. The propionibacterium Propionibacterium freudenreichii has been most often investigated (rarely other bacteria, e.g. Bacillus megaterium (Reference Kysil61) and Acetobacter pasteurianus (Reference Bernhardt, Zhu and Schütz532)) for fortification by fermentation during food processing; and various substrates have been fermented, including cereals (e.g. wheat, barley, rye, oat, rice, sorghum and millet), pseudocereals (e.g. buckwheat, quinoa and amaranth), legumes (e.g. faba bean, soy bean and lupin beans), whey, soy milk, sunflower seed milk, cabbage, ground elder and black tea(Reference Kysil61,Reference Edelmann, Chamlagain and Santin386,Reference Chamlagain420,Reference Calvillo, Pellicer, Carnicer and Planas450,Reference Chamlagain, Deptula and Edelmann533Reference Coelho, de Almeida and do Amaral550) . The usefulness of lactic acid bacteria, such as Lactobacillus plantarum, Lactobacillus reuteri and Lactobacillus rhamnosus, for in situ fortification with vitamin B12 is questionable. It is not clear which, if any, of these strains synthesises true cobalamin and not merely pseudocobalamin. The ability of lactic acid bacteria to produce vitamin B12 is, in fact, usually evidenced by a microbiological assay that does not distinguish between cobalamin and pseudocobalamin and is not verified by a reliable analytical method for structure elucidation and/or by a genetic analysis confirming the presence of the whole biosynthetic pathway in the genome of a particular micro-organism(Reference Watanabe, Bito and Koseki79,Reference Masuda, Ide and Utsumi303,Reference Vincenti, Bertuzzo and Limitone372,Reference Deptula, Chamlagain and Edelmann536,Reference Xie, Coda and Chamlagain539,Reference Hugenschmidt, Schwenninger, Gnehm and Lacroix545,Reference Capozzi, Russo and Duenas547,Reference Gu, Zhang and Song551Reference De Angelis, Bottacini and Fosso563) . The practical importance of vitamin B12 biofortification strategies, including in situ methods, is so far low, if any, compared with fortification (i.e. external addition of cobalamin to foods). However, it is a promising way to provide the vitamin to consumers and requires further research.

Pharmacokinetics and homoeostasis

Absorption

In humans, the selective absorption of vitamin B12 is a multi-step process (Figure 2). The bioavailability depends on the individual’s gastrointestinal absorption capacity and, in terms of food sources, on the amount and type of protein consumed. Indeed, vitamin B12 ingested through food appears to have varying rates of absorption(Reference O’Leary and Samman5,564) . In general, the bioavailability of the vitamin from the usual diet is assumed to be about 50% (depending on the dietary source, the amount of cobalamin ingested, the ability to release cobalamin from food and the proper functioning of the intrinsic factor system), but lower from sources containing high amounts, e.g. from liver, due to saturation of the active absorption process(63,Reference Allen66,Reference Sobczyńska-Malefora, Delvin and McCaddon81,Reference Watanabe84,88,Reference Allen92,Reference Allen, Miller and de Groot93,Reference Doets, Szczecińska and Dhonukshe-Rutten98,Reference Stabler, Bernadette, Marriott and Yates152,Reference Yates565) .

Fig. 2. Absorption of vitamin B12 via the IF pathway: Dietary protein-bound vitamin B12 can bind to transcobalamin I (TCI) only after its release mediated by pepsin and hydrochloric acid produced by the gastric mucosa. In the duodenum, TCI is degraded by pancreatic proteases and free cobalamin binds to intrinsic factor (IF). The IF–cobalamin complex is absorbed in the distal ileum by receptor-mediated endocytosis enabled by cubilin with participation of other protein(s), e.g. amnionless (AMN). IF is degraded in the lysosome and released cobalamin enters the cytoplasm likely by use of the transmembrane protein LMBD1. The precise mechanism of vitamin B12 efflux from enterocytes into the circulation is not yet well described. It appears to be mediated by several exporters; one of them is multidrug resistance protein 1 (MRP1, shown in teal colour).

Vitamin B12 is bound to proteins in food and is available for absorption only after releasing by pepsin and hydrochloric acid produced by the gastric mucosa. Subsequently, it binds to transcobalamin I (TCI) belonging to haptocorrins (HC)/R binders. Due to this classification, it is sometimes simply referred as haptocorrin. It is a glycoprotein that is found in saliva and gastric fluids and, inter alia, in blood serum. TCI has a high affinity for both B12 and for its analogues. In the duodenum, TCI is degraded by pancreatic proteases and free cobalamin binds to intrinsic factor (IF), a glycoprotein that is secreted by gastric parietal cells after a meal. There is high homology between IF and TCI, and both of them bind one molecule of vitamin B12 (Reference Kozyraki and Cases566). Cobalamin binds to IF with a higher affinity in a more alkaline environment; hence, in the stomach, where the pH is acidic, IF has a very low affinity for vitamin. This glycoprotein is much more specific for B12 binding than TCI and has limited affinity for cobalamin analogues. The IF–cobalamin complex is absorbed in the distal ileum by receptor-mediated endocytosis enabled by cubilin with participation of other protein(s), e.g. amnionless (AMN). IF is degraded in the lysosome, and released B12 enters the cytoplasm likely by use of the transmembrane protein LMBD1. The precise mechanism of vitamin B12 efflux from enterocytes into the circulation is not yet well described. It appears to be mediated by several exporters; one of them is multidrug resistance protein 1 (MRP1)(Reference O’Leary and Samman5,Reference Green, Allen and Bjørke-Monsen260,Reference Kozyraki and Cases566) .

The normal mechanism of absorption of orally administered vitamin B12, via the IF pathway, is readily saturated. While approximately 70% of vitamin B12 is absorbed from doses of 0.1–0.5 μg, it decreases to 56% at 1 μg, to 16% at 10 μg and to 3% for doses 25–50 μg(Reference Smith, Warren and Refsum6). High oral doses (100–100 000 μg) are absorbed passively, but the extent reaches only about 1% of the ingested dose(Reference Berlin, Berlin and Brante567).

Transport

In the bloodstream, the majority of B12 (80%) and all cobalamin analogues are bound to TCI, which, thanks to its relatively long biological half-life of 10 d, forms a circulating supply of vitamins in the body(Reference Fedosov and Stanger568). Of the total B12, 20–30% is carried by transcobalamin II (TCII), a non-glycosylated protein(Reference Smith, Warren and Refsum6). TCII binds physiological forms of vitamin B12, while TCI also binds B12 analogues. TCII and TCI deliver vitamin B12 to peripheral tissues and liver, respectively(Reference Shipton and Thachil569). The TCII–cobalamin complex binds in the presence of calcium to its receptor, a transmembrane, highly glycosylated protein CD320 (8D6A) containing two low-density lipoprotein-receptor class A domains. The receptor is selective to cobalamin–TCII, and neither TCI nor IF binds to it(Reference Quadros, Nakayama and Sequeira570). After endocytosis, the complex enters the lysosome, where TCII is degraded and free B12 is exported to the cytosol by use of the ATP-binding cassette transporter ABCD4. Presence of another membrane lysosomal protein LMBD1 is also necessary (Reference Smith, Warren and Refsum6,Reference Watkins and Rosenblatt571) . A third specific vitamin B12 transport protein found in human serum, transcobalamin III (TCIII), is also reported in the literature(Reference Bloomfield and Scott572Reference Wickramasinghe, England, Saunders and Down576). Like TCI, TCIII is a glycoprotein and, in addition, these two transcobalamins are immunologically identical (Reference Burger, Schneider, Mehlman and Allen573,Reference Fràter-Schröder, Hitzig and Bütler574) .

Reabsorption and excretion

Vitamin B12 is secreted into the bile, and a part is reabsorbed by the enterohepatic circulation through ileal receptors that require IF(Reference Kozyraki and Cases566,Reference Herrmann and Obeid577) . Cobalamin is excreted in the faeces, which consists of unabsorbed biliary vitamin B12, vitamin B12 from gastrointestinal cells and secretions, and that synthesised by bacteria in the colon. When the vitamin B12 is found in the excess in the circulation, it outreaches the binding capacity of TCII, and it is also excreted in the urine(Reference O’Leary and Samman5). However, it is partially reabsorbed in the kidney by the transcobalamin II receptor megalin (Lrp2)(Reference Kozyraki and Cases566). Further losses of vitamin B12 occur through the skin and metabolic reactions(564).

Storage of cobalamin in the human body

Cobalamin, unlike other water-soluble vitamins, is stored in the human body. Most adults have stores of up to 5 mg. The liver is the main reservoir of this vitamin, and it stores normally up to one half of the total amount of the vitamin(Reference Herrmann and Obeid577,Reference Manzanares and Hardy578) . Smaller amounts of accumulated cobalamin can also be found in the kidneys and brain, and the circulating supply of TCI-bound vitamin B12 in plasma cannot be neglected(Reference Herrmann and Obeid577).

Physiological function

Vitamin B12 is essential for human metabolism, production and regeneration of carbohydrates, fats and proteins, as well as for the proper development of erythrocytes and the central nervous system. Only two forms are biologically active: methylcobalamin and adenosylcobalamin(564,Reference Kozyraki and Cases566) . The former is a cofactor for methionine synthase and the latter for l-methylmalonyl-CoA mutase (Figure 3). These B12-mediated reactions are facilitated by the ability of the cobalt ion to change its oxidation states, Co (I), Co (II) and Co (III) (Figures 4 and 5)(Reference Kräutler579,Reference Mascarenhas, Gouda, Ruetz and Banerjee580) . Co (I) is unstable and acts as a supernucleophile. The corrin ring helps to stabilise this form(Reference Smith, Warren and Refsum6).

Fig. 3. Physiological function of vitamin B12 and its connection with folate metabolism: (A) Together with folic acid (vitamin B9), methylcobalamin as a cofactor for the enzyme methionine synthase is necessary for the formation of methionine. During the reaction, the methyl group is transferred from methyltetrahydrofolate (CH3-THF) to homocysteine by the enzyme; the resulting tetrahydrofolate can be then converted to methylenetetrahydrofolate (CH2=THF), the form required for de novo thymidine synthesis. (B) In the conversion of methylmalonyl-coenzyme A to succinyl-coenzyme A, B12 is involved in its active form adenosylcobalamin as a cofactor of the enzyme methylmalonyl-coenzyme A mutase. The resulting succinyl-coenzyme A is a major mediator of the tricarboxylic acid (TCA) cycle; CoA, coenzyme A; DHF, dihydrofolate; THF, tetrahydrofolate.

Fig. 4. Formation of methylcobalamin: The highly nucleophilic cob(I)alamin reacts with a methylating agent to form methylcobalamin. Modified in ChemDraw, version 20.0 on the basis of publication of Kräutler(Reference Kräutler579).

Fig. 5. Formation of adenosylcobalamin: Adenosylcobalamin functions as a reversible source of the 5′-deoxyadenosyl radical, this reaction produces cob(II)alamin. Modified in ChemDraw, version 20.0 on the basis of publication of Kräutler(Reference Kräutler579).

Methionine synthase

Methionine synthase is a cytoplasmic enzyme requiring both vitamin B12 and vitamin B9. It converts homocysteine into methionine by transfer of a methyl group. The donor of the methyl group is methylcobalamin, which is subsequently recovered by transfer of one carbon unit from methylenetetrahydrofolate (vitamin B9). It need not be emphasised that mutation in methionine synthase or derangement in vitamin B12 physiology leads to hyperhomocysteinaemia(Reference Watkins, Ru and Hwang581). High plasma homocysteine levels are considered vasculotoxic and neurotoxic, but the relationship between human diseases, homocysteine levels and supplementation by B vitamins to decrease the homocysteine levels it is still a matter of debate(Reference Kim, Kim, Roh and Kwon582,Reference McCaddon and Miller583) . Furthermore, the formation of the essential amino acid methionine allows several methylation reactions necessary for the synthesis of nucleotides for DNA/RNA and proteins. Failure of this step can be observed especially in rapidly multiplying cells, such as erythrocytes or enterocytes, and can affect several processes including the growth of vascular endothelial cells or the production of noradrenaline, which is involved in both stress response and cardiovascular system function(Reference Allen, Miller and de Groot93,Reference Kozyraki and Cases566,Reference Allen584) .

Methylmalonyl-coenzyme A mutase

The mitochondrial enzyme methylmalonyl-coenzyme A mutase is involved in the catabolism of odd-chain fatty acids, some branched-chain amino acids and cholesterol to form succinyl-coenzyme A. A defect in this response is thought to be involved in several neurological manifestations of vitamin B12 deficiency, including movement disorders, seizures and mental retardation(Reference Allen, Miller and de Groot93,Reference Kozyraki and Cases566,Reference Allen584) .

Laboratory assessment of vitamin B12 status

Vitamin B12 status can be assessed by measuring the serum/plasma B12 level, which is the sum of B12 TCI-bound (holohaprocorrin) and TCII-bound (holotranscobalamin); serum holotranscobalamin (holoTC) concentration; serum/plasma methylmalonic acid (MMA) concentration; and total serum homocysteine (tHcy) concentration(4,Reference Harrington585) . However, no single laboratory marker is suitable for assessing B12 status in all patients. Combinations, such as multiple markers or sequential assay selection algorithms(Reference Harrington585) or the calculations(Reference Fedosov586), that combine the single B12 diagnostic indicators, such as combined indicator (cB12), which uses all four most commonly available markers: cB12 = log10[(holoTC × total serum B12 level)/(MMA × tHcy)] − (age factor), are usually employed for more precise determination of vitamin B12 levels(Reference Harrington585,Reference Fedosov, Brito and Miller587) . Analytical methods used for the measurement of vitamin B12 in serum/plasma and other biological fluids are summarised in Table 2.

Table 2. Summary of analytical methods for the assessment of vitamin B12 in biological fluids

Determination of total serum/plasma cobalamin

Serum or plasma B12 levels provide information on the long-term B12 status and liver stores. Recent intake has no particular effect. This marker is not very sensitive or specific, leading to a false positive and negative diagnosis. A serum level of vitamin B12 below 148 pM with symptoms is a strong indicator of deficiency, but symptoms might be present even with serum levels above this value(Reference Wolffenbuttel, Owen, Ward and Green2). Conditions that increase TCI levels such as chronic granulocytic leukaemia, autoimmune lymphoproliferative syndrome, alcoholism, liver disease and cancer will also elevate vitamin B12 levels(Reference Allen, Miller and de Groot93).

Determination of holotranscobalamin

Serum holoTC levels determine the amount of the physiologically active form of cobalamin bound to TCII. As aforementioned, holoTC accounts for 20–30% of total B12 serum levels. This method is the most sensitive to recent intake with a response within few hours. Hence, this marker can be increased even when body reserves are low. HoloTC levels are, however, also increased in patients with renal impairment(Reference Allen, Miller and de Groot93).

Determination of methylmalonic acid

Serum MMA is the most sensitive biomarker of B12 status. It is a good indicator of liver stores, and it reflects the utility of vitamin B12 for metabolic functions, i.e. methylmalonyl CoA mutase activity. MMA as a by-product of methylmalonyl CoA metabolism increases in B12 deficiency. When total serum B12 is <287 pM, its concentrations increase. MMA metabolite is affected neither by folate nor other B vitamins, but it increases with renal dysfunction, so serum creatinine should be measured, especially in the elderly. MMA levels increase with aging, especially after age 70, and neither lower intake nor impaired renal function fully explains why this occurs. Higher serum concentrations of MMA occur also with an overgrowth of intestinal bacteria that produce propionic acid. Conversely, antibiotic treatment may lower the level(Reference Allen, Miller and de Groot93).

Determination of total serum homocysteine

The biomarker tHcy is not specific for B12 levels. It reflects the availability of B12 for metabolic functions, i.e. methionine synthase activity. Concentrations increase when total serum B12 is <300 pM, but also in deficiency of folates, riboflavin and vitamin B6, as well as with renal insufficiency and hypothyroidism(Reference Allen, Miller and de Groot93).

Combination of biomarkers

The combination of all four biomarker values (serum B12 levels, holoTC, tHcy and MMA) or the cB12 mathematical model using ‘four-biomarker’ analysis appears to be the most accurate for determining B12 levels, but in some cases is unbearably expensive(Reference Allen, Miller and de Groot93). A study by Fedosov et al.(Reference Fedosov, Brito and Miller587) showed that the cB12 model using analysis of ‘three-biomarker‘or ‘two-biomarker’ estimates the level of B12 within acceptable error limits compared with the analysis of ‘four-biomarker’. The combination of holoTC, MMA and total serum B12 levels appears to be the best in the ‘three-biomarker’ analysis. Further quality analysis is achieved by omitting total serum B12 or holoTC from the ‘four-biomarker’ analysis. In terms of ‘two-biomarker’ analysis, the smallest error was observed when holoTC was used in conjunction with the MMA assay. The article by Fedosov et al.(Reference Fedosov, Brito and Miller587) also provides formulas for cB12 for cases where one or two biomarkers are missing.

Identification of aetiology of vitamin B12 deficiency

A formerly used Schilling test was withdrawn mainly due to concerns of bovine spongiform encephalopathy transmission via its animal-derived intrinsic factor(Reference Harrington585,Reference Carmel588) . Another way to detect a deficiency caused by impaired absorption is the CobaSorb test, which uses holoTC as a sensitive marker of the recent intake of B12. The test consists of taking blood samples before and 1–2 d after taking an oral dose of 9 µg of cyanocobalamin three times a day and measuring the increase in holoTC(Reference von Castel-Roberts, Morkbak and Nexo589). This method can detect holoTC elevation of ≥10 pM only if the holoTC baseline was <75 pM. Under these conditions, both sensitivity and specificity are considered excellent(Reference Hvas, Morkbak and Nexo590). Hardlei et al. improved this method. After stating that a major part of the oral test dose of cyanocobalamin is absorbed without modification, it was suggested that the capacity to absorb vitamin B12 can be evaluated by measuring TC-cyanocobalamin before and after administration of the test dose of cyanocobalamin. C-Cobasorb, as this method is named, has a higher specificity than the previously mentioned CobaSorb(Reference Hardlei, Mørkbak and Bor591). Therefore, if holoTC baseline is >65 pM, it is recommended to use the C-CobaSorb assay for the assessment(Reference Nexo and Hoffmann-Lücke592). Other laboratory tests usable for determination of the cause of B12 deficiency are tests based on plasma IF antibodies, plasma gastrin and pepsinogen I, and plasma parietal cell antibodies(Reference Harrington585,Reference Alonso Nr and Salinas593) .

Cobalamin deficiency

Subclinical and clinical cobalamin deficiency

Low levels of vitamin B12 can be divided into four stages. The first two stages represent depletion, while the second two represent deficiency. Stage I is low serum vitamin B12; stage II is low stores in cells; stage III is biochemical deficiency; and stage IV is clinical deficiency with overt manifestations(Reference Herbert594). Other classification divides vitamin B12 deficiency into clinical and subclinical forms. Subclinical cobalamin deficiency involves mild biochemical changes without clinical manifestations. Clinical deficiency usually results from severe, persistent malabsorption, while dietary insufficiency, intermittent or partial malabsorption can usually merely induce only subclinical cobalamin deficiency(Reference Carmel595).

At the cellular and molecular level, vitamin B12 deficiency manifests itself in one or both forms of coenzyme B12 (methylcobalamin and adenosylcobalamin). Methylcobalamin deficiency leads to impaired nucleotide synthesis and methylation, and adenosylcobalamin deficiency disrupts the metabolism of methylmalonate, which is derived from the catabolism of odd-chain fatty acids and ketogenic amino acids(Reference Allen, Miller and de Groot93,Reference Green, Allen and Bjørke-Monsen260) . This can also be the key to understanding the mechanisms responsible for clinical manifestations.

Causes of vitamin B12 deficiency and diagnosis

B12 deficiency can be caused by several pathophysiological processes. They can affect both B12 supply and demand, and they can occur at any time during human life. Specifically, cellular B12 deficiency could be due to insufficient intake and/or bioavailability including malabsorption, chemical inactivation(Reference Flippo and Holder3) or disruption of B12 transport in the blood or intracellular uptake and metabolism. Also, some diseases, medications and bacterial overgrowth are related to B12 deficiency(Reference O’Leary and Samman5,Reference Green, Allen and Bjørke-Monsen260,Reference Herrmann and Obeid577) . It can also occur in people with increased demands, such as during major bleeding, and in pregnant or breastfeeding women, and this can impact their infants as well(Reference Green, Allen and Bjørke-Monsen260,Reference Herrmann and Obeid577) .

People from developing countries and those who do not voluntarily consume animal products because of their religion, culture or personal attitude may suffer from B12 deficiency(Reference Green, Allen and Bjørke-Monsen260,Reference Herrmann and Obeid577) . In terms of new vegans, the symptoms of vitamin B12 deficiency may not appear for several years owing to the large reserves in the human body and reabsorption of the vitamin(Reference Shipton and Thachil569,Reference Herrmann and Obeid577) . Lacto- and lacto-ovo-vegetarian diet could be sufficient thanks to milk and eggs that contain a low amount of cobalamin(Reference Herrmann and Obeid577). Metabolic and clinical signs of cobalamin deficiency have been reported in neonates from strict vegetarian mothers or in breastfed infants by cobalamin-deficient mothers(Reference Kozyraki and Cases566,Reference Higginbottom, Sweetman and Nyhan596,Reference Monsen, Ueland and Vollset597) . To avoid the risk of deficiency, they can obtain cobalamin from fortified foods (see section Prevention by fortification of food) or food supplements(Reference O’Leary and Samman5,Reference Green, Allen and Bjørke-Monsen260) .

Furthermore, the deficiency occurs because of cobalamin malabsorption. In the case of pernicious anaemia (chronic atrophic gastritis type A), cobalamin cannot be absorbed because of the lack of IF. It is a consequence of autoimmune gastritis when the parietal cells producing IF and hydrochloric acid in the stomach are destroyed. Chronic atrophic gastritis type B, which is related to persistent infection with Helicobacter pylori, may also impair vitamin B12 absorption. Both are due to disturbance of the pH of the stomach and the inability to release vitamin B12 from food proteins(Reference Green, Allen and Bjørke-Monsen260,Reference Kozyraki and Cases566,Reference Herrmann and Obeid577) . Proton pump inhibitors, H2-receptor antagonists and antacids also suppress the gastric acidity and, hence, similarly impair vitamin B12 absorption(Reference Green, Allen and Bjørke-Monsen260,Reference Shipton and Thachil569,Reference Herrmann and Obeid577,Reference Manzanares and Hardy578) . Diseases affecting the small intestine such as coeliac disease, Crohn’s disease or ulcerative colitis are connected with B12 malabsorption due to villous atrophy and mucosal injury or recurrent diarrhoea(Reference O’Leary and Samman5,Reference Green, Allen and Bjørke-Monsen260,Reference Shipton and Thachil569,Reference Herrmann and Obeid577) . Last but not least, cobalamin malabsorption is associated with gastric, post-gastric or ileal resection, or pancreatic insufficiency or pancreatectomy that cause inability of B12 release from HC binding(Reference O’Leary and Samman5,Reference Green, Allen and Bjørke-Monsen260,Reference Shipton and Thachil569) . Another disease associated with low levels of cobalamin is HIV infection. Plasma concentrations decrease with disease progression(Reference Allen66). In addition, older people are at risk of malabsorption not only because of the various diseases and medications they take, but also because of the natural aging process. Several age-related physiological factors can adversely affect the absorption of the vitamin from the intestine(Reference Herrmann and Obeid577).

In addition to the above-mentioned drugs, low levels of vitamin B12 are also associated with oral contraceptives(Reference Berenson and Rahman598), metformin, cholestyramine, colchicine, several antibiotics, and drugs such as p-aminosalicylic acid(Reference Green, Allen and Bjørke-Monsen260,Reference Manzanares and Hardy578) and in elderly patients also with ACE inhibitors(Reference Tal, Shavit, Stern and Malnick599). In some, the mechanism is known as with cholestyramine that may bind intrinsic factor, while colchicine, several antibiotics, and antituberculotic drug p-aminosalicylic acid, may act as inhibitors of intrinsic factor-B12 endocytosis(Reference Green, Allen and Bjørke-Monsen260).

In terms of the chemical inactivation, the anaesthetic gas nitrous oxide irreversibly oxidates cobalt. This oxidation is, however, relevant solely the enzyme methionine synthase(Reference Flippo and Holder3,Reference Green, Allen and Bjørke-Monsen260,Reference Chanarin600) . On the other hand, adenosylcobalamin is not changed. The reason is that mechanisms of the catalytic reaction differ between methionine synthase and methylmalonyl-coenzyme A mutase particularly in relation to changes in the oxidation state of the central cobalt ion. During methionine synthesis, cob(I)alamin intermediate, which is susceptible to oxidation, is formed, whereas during methylmalonyl-succinyl coenzyme A transformation, cobalt is never reduced to Co(I) and, thus, not susceptible to N2O-induced oxidation. (Reference Mascarenhas, Gouda, Ruetz and Banerjee580,Reference Chanarin600) .

Moreover, there are some rare genetic defects causing impairment in absorption, transport, metabolism or utilisation of cobalamin. Hereditary IF deficiency caused by recessive mutations in the IF gene (CBLIF) is an inherited disorders of vitamin B12 absorption associated with congenital pernicious anaemia. Imerslund–Gräsbeck syndromes 1 and 2 (also known as hereditary megaloblastic anaemia 1 and 2) are associated with selective vitamin B12 malabsorption in the ileum; there is a defect in transport of cobalamin into the enterocytes due to mutation in cubilin (gene CUBN) and amnionless (gene AMN), respectively. With respect to the physiological function of TCI (gene TCN1) and TCII (gene TCN2), it is clear that congenital deficiency of at least one of them leads to defective absorption as well as transport into cells. In addition, mutations in TCII receptor CD320 are known. There is also a group of historically so-called cobalamin mutant diseases cblA, cblB, cblC, cblD, cblE, cblF, cblG, cblJ, cblK and cblX. These diseases affect different processes associated with vitamin B12: (1) efflux from lysosomes to cytosol (cblF – gene LMBRD1, cblJ – gene ABCD4); (2) intracellular vitamin B12 trafficking via chaperone MMACHC (cblC, /gene MMACHC/), silencing its gene (‘epi-cbl C’, gene PRDX1), or proteins regulating its expression – cblX /gene HCFC1 on X chromosome/ and cblK /gene ZNF143/; (3) likely transport to methionine synthase – cblD /gene MMADHC/; (4) metabolism by methionine synthase (cblG, methione synthase, gene MTR) or methionine synthase reductase (cblE, gene MTRR); (5) synthesis of adenosylcobalamin by ATP:cobalamin adenosyltransferase /cblB, gene MMAB/ and transport of produced adenosylcobalamin to methylmalonyl CoA mutase /cblA, gene MMAA/. Methylmalonyl CoA mutase deficiency (gene MMUT) is also known(Reference Kozyraki and Cases566,Reference Watkins and Rosenblatt571,Reference Herrmann and Obeid577,Reference Watkins, Ru and Hwang581,Reference Rosenblatt and Cooper601) .

The reference intervals of the individual biomarkers as well as their values indicating B12 deficiency are given in Table 3 (Reference Green, Allen and Bjørke-Monsen260,Reference Fedosov, Brito and Miller587) . To determine the aetiology of vitamin B12 deficiency, the (C-)CobaSorb assay or plasma IF antibodies, plasma gastrin and pepsinogen I, and plasma parietal cell antibodies (Reference Harrington585,Reference Alonso Nr and Salinas593) can be used, as mentioned above.

Table 3. The reference intervals of the individual biomarkers, values indicating transitional status and B12 deficiency

B12, total serum B12 concentrations; cB12, combined indicator of vitamin B12 status.

Clinical manifestations

Clinical deficiency is manifested by haematological, neurological and neuropsychiatric symptoms(Reference Wolffenbuttel, Owen, Ward and Green2,Reference Green, Allen and Bjørke-Monsen260,Reference Stabler441,Reference Shipton and Thachil569,Reference Herrmann and Obeid577,Reference Dali-Youcef and Andrès602Reference Reynolds604) . No specific correlation between haematological and neurological symptoms has been demonstrated, so patients with neurological manifestations may not have any haematological abnormalities and vice versa(Reference Allen, Miller and de Groot93,Reference McCaddon605) .

Haematological manifestations are usually but not always connected with macrocytic or megaloblastic anaemia that is characterised by enlarged but less numerous erythrocytes, and by hypersegmented neutrophils. It is the result of disruption of DNA synthesis. In B12 deficiency, the recovery of tetrahydrofolate is disrupted. This limits the supply of folate for the synthesis of thymidylate, purine nucleotides and, subsequently, DNA. The haematopoietic system is particularly affected because blood cells are cells with a rapid turnover. DNA synthesis in bone marrow blood cell precursors is stopped, which prevents mitosis but allows cytoplasmic maturation(Reference Allen, Miller and de Groot93,Reference Green, Allen and Bjørke-Monsen260) . Because other haematopoietic cells are also affected, it can lead to isolated thrombocytopenia and neutropenia and even pancytopenia with impairment of cellular and humoral immunity(Reference Allen, Miller and de Groot93,Reference Green, Allen and Bjørke-Monsen260,Reference Stabler441,Reference Shipton and Thachil569,Reference Dali-Youcef and Andrès602) .

Cobalamin deficiency appears to be more common in patients with a number of chronic neurological and neuropsychiatric diseases. Clinical manifestations of vitamin B12 deficiency can include several CNS symptoms (peripheral neuropathy, subacute combined degeneration of the spinal cord, paraesthesia, ataxia, abnormal reflexes, bowel and bladder incontinence, erectile dysfunction, stroke, optic atrophy, orthostatic hypotension, dementia, multiple sclerosis, Alzheimer’s disease, parkinsonian syndromes, depression, mania, irritability, paranoia, delusions, psychosis, delirium), but also atherosclerosis. However, it is still not clear whether there is a direct causality(Reference Stabler441,Reference Shipton and Thachil569,Reference Herrmann and Obeid577,Reference Dali-Youcef and Andrès602,Reference Lachner, Steinle and Regenold603) . There are several theories that link cobalamin deficiency with neurological and neuropsychiatric problems such as (a) disorders of the formation of monoamine neurotransmitters, because cobalamin and folate are essential for the production of tetrahydrobiopterin that is required for monoamine synthesis(Reference Hutto606); (b) DNA synthesis disturbances due to deficiency of methyl donors(Reference Reynolds604); (c) hyperhomocysteinaemia and its discussed vasculotoxic and neurotoxic effects(Reference McCaddon and Miller583,Reference Reynolds604,Reference Obeid, McCaddon and Herrmann607,Reference Norbert Goebels and Michael Soyka608) ; (d) demyelination due to a lack of S-adenosylmethionine as a consequence of inhibition of homocysteine methylation to methionine(Reference Reynolds604,Reference Norbert Goebels and Michael Soyka608,Reference Metz609) ; (e) inhibition of the methylmalonyl-succinyl CoA pathway causes disruption of odd–chain fatty acid metabolism and may also lead to demyelination(Reference Reynolds604,Reference Metz609) .

In addition, there is a higher risk for infants born to B12-deficient women to develop neural tube defects with impairment of psychomotor function and brain development, anaemia and growth disorders. Neural disturbances may be irreversible(Reference Niklewicz, Smith and Smith244,Reference Finkelstein, Layden and Stover610) .

Prevention by fortification of food

Fortification of foods with vitamin B12 is primarily intended for populations at risk for its deficiency. The highest prevalence of deficiency occurs in people who have a low dietary intake of the vitamin and in those, primarily the elderly, suffering from malabsorption of food-bound cobalamin, whose vitamin B12 status can be poor despite intakes often appearing to be adequate. As already discussed, vegetarian diets also represent a clear risk factor for vitamin B12 deficiency, as those diets have become very popular in the past few decades, especially in developed countries, because of their potential health benefits but also due to ethical and environmental issues. The diets of populations in low- and middle-income countries are typically low in animal-source foods because of their relatively high cost, lack of availability, and/or cultural and religious reasons(Reference Allen66,Reference Sobczyńska-Malefora, Delvin and McCaddon81,Reference Jiang, Christian and Khatry611Reference Thankachan, Rah and Thomas613) . Reduced acid production in the stomach associated with atrophic gastritis does not interfere with the absorption of the free crystalline vitamin present in fortified foods or supplements because intrinsic factor is still secreted(63,Reference Allen66,Reference Sobczyńska-Malefora, Delvin and McCaddon81,Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Allen, Miller and de Groot93,Reference Truswell137,Reference Stabler, Bernadette, Marriott and Yates152,Reference Tucker, Olson and Bakun199,Reference Winkels, Brouwer and Clarke200,Reference Melina, Craig and Levin205,Reference Green, Allen and Bjørke-Monsen260,Reference Carmel455,Reference Yates565,Reference Allen, Rosenberg, Oakley and Omenn612,Reference Allen614Reference Oakley623) . Fortification with cobalamin has also been recommended in foods fortified with folate because of concerns that excess intake of folic acid alone by people with a low vitamin B12 status, particularly in the elderly, may delay the diagnosis of cobalamin deficiency. High levels of folate may mitigate (‘mask’) symptoms of anaemia caused by cobalamin deficiency and cannot correct nervous system damage, allowing or even accelerating its progression in persons with unrecognised and untreated cobalamin deficiency. Significance of this phenomenon has been an issue of persistent debate(63,Reference Winkels, Brouwer and Clarke200,Reference Agnoli, Baroni and Bertini242,Reference Green453,Reference Refsum and Smith615Reference Blacher, Czernichow and Raphaöl619,Reference Oakley623Reference Miller, Garrod and Allen656) . Whether the concerns are substantiated or not(Reference Obeid, Heil and Verhoeven108,Reference Carmel456,Reference Allen, Rosenberg, Oakley and Omenn612,Reference Mills, Molloy and Reynolds627,Reference Brouwer and Verhoef639,Reference Berry657Reference Crider, Bailey and Berry666) , cobalamin deficiency should be prevented in any case, and fortification with both folate and cobalamin would reduce possible risks. As of 2022, there are twenty-five countries in Africa, Central and South America, and Asia having mandatory fortification of wheat flour, maize flour or rice with vitamin B12 (Burundi, Cameroon, Chad, Ethiopia, Ghana, Kenya, Liberia, Malawi, Mozambique, Nigeria, Rwanda, Tanzania, Uganda, Zimbabwe, Costa Rica, Cuba, Guatemala, Nicaragua, Panama, Peru, Afghanistan, Jordan, Palestine, Uzbekistan and Vietnam)(667Reference Bobrek, Broersen and Aburto670). In addition, food is fortified with the vitamin on a voluntary basis in many other countries including, e.g., the United States, Brazil, India, Bangladesh, Myanmar, Indonesia, Sudan, Sierra Leone and countries of the European Union(Reference Zeuschner, Hokin and Marsh224,444,449,Reference Calvillo, Pellicer, Carnicer and Planas450,Reference Hopkins, Gibney and Nugent461,Reference Titcomb and Tanumihardjo521,667,668,Reference Muthayya, Hall and Bagriansky671Reference Laird, O’Halloran and Carey674) . Foods fortified with vitamin B12 include cereal-based foods, meat substitute products (such as tofu), milk substitutes (such as soy-based milk and yoghurt alternatives), nutritional yeast, fruit juices, multivitamin sweets and milk powder infant foods(Reference Watanabe, Yabuta, Tanioka and Bito85,Reference Allen, Miller and de Groot93,Reference Stabler, Bernadette, Marriott and Yates152,Reference Tucker, Olson and Bakun199,Reference Sebastiani, Herranz Barbero and Borrás-Novell216,Reference Zeuschner, Hokin and Marsh224,Reference Chandra-Hioe, Lee and Arcot256,Reference Marczykowski and Breidenassel259,444,449,Reference Allen, Rosenberg, Oakley and Omenn612,Reference Thankachan, Rah and Thomas613,Reference Garrod, Buchholz and Miller636,Reference Engle-Stone, Nankap and Ndjebayi669,Reference Muthayya, Hall and Bagriansky671,Reference Sirohi, Pundhir and Ghosh672,Reference Dunn, Jain and Klein675Reference Madhari, Boddula and Ravindranadh679) . Attractive strategies for fortification have been investigated, such as fortification of milk (Reference Dhonukshe-Rutten, van Zutphen and de Groot616,Reference Sanchez, Albala and Lera621) , yoghurt(Reference Melo, Ng and Tsang680), tea (Reference Vora, Alappattu and Zarkar681,Reference Vora and Antony682) , fruit and vegetable juices(Reference Bajaj and Singhal388), ready-to-blend fresh-cut fruit/vegetable mix(Reference Artés-Hernández, Formica-Oliveira, Artés and Martínez-Hernández683), mineral water(Reference Tapola, Karvonen, Niskanen and Sarkkinen684), sugar cubes(Reference Bajaj and Singhal438), salt (Reference Modupe and Diosady391,Reference Vinodkumar and Rajagopalan439) , dried soup(Reference Sanchez, Albala and Lera621) and toothpaste(Reference Zant, Awwad and Geisel685,Reference Siebert, Obeid and Weder686) .

Therapy

The first-line treatment in patients with risk factors for the vitamin deficiency is prevention of this deficiency and/or supplementation with vitamin B12. Treatment should be started as soon as possible(Reference Green, Allen and Bjørke-Monsen260). Parenteral therapy or high-dose oral administration can be used to treat vitamin B12 deficiency if due to inadequate dietary intake. In diagnosed vitamin B12 deficiency, intramuscular administration should be preferred as the effect or oral treatment is lower based on current knowledge(Reference Wolffenbuttel, Owen, Ward and Green2). Intramuscular injections of 1000 μg of cyanocobalamin or hydroxocobalamin are given daily or every other day for at least one week followed by weekly injections for at least one month. Thereafter, they are reduced to a dose of 1000 μg of cyanocobalamin monthly. Injections of hydroxocobalamin can be given each 3 months after the initial intensive therapy. Alternatively, subcutaneous administration is recommended in cases of contraindications of intramuscular application, e.g. when a patient is treated with anticoagulants. Oral treatment consists of 1000–2000 μg of cyanocobalamin orally per day. In some countries, also sublingual and intranasal vitamin B12 is available, but their clinical profit has not yet been clearly established(Reference Wolffenbuttel, Owen, Ward and Green2,Reference Green, Allen and Bjørke-Monsen260,Reference Stabler441,Reference Dali-Youcef and Andrès602) . The development of nanoparticles for oral vitamin B12 administration able to overcome the IF-absorption pathway can enrich the palette of current treatment modalities in the future(Reference Fidaleo, Tacconi and Sbarigia452).

The duration of treatment depends on the cause and clinical manifestations of the deficit. Blood count usually recovers within 2 months, and neurological signs correct or improve within 6 months(Reference Stabler441). However, in patients with severe neurological disorders, improvement usually will be weak after 1 year of adequate therapy. These patients may suffer from permanent impairments(Reference Green, Allen and Bjørke-Monsen260). In case of malabsorption, treatment continues even after the symptoms have disappeared; it is usually a life-long therapy. Both routes of administration (including self-injection at home) can be used(Reference Wolffenbuttel, Owen, Ward and Green2). Selection of the route of administration usually depends on patient preferences and the compliance. As mentioned above, other less studied routes of administration (sublingual, nasal or even transdermal) are available, but they are also more expensive(Reference Stabler441,Reference Carmel455) . Regarding dietary deficiency, it is recommended to take at least 6 μg/d after the symptoms have disappeared and the body’s vitamin B12 levels have been restored. In infants, treatment is usually started with intramuscular injections of 250–1000 μg cyanocobalamin or hydroxocobalamin daily, then until recovery once a week, followed by an oral administration of 1–2 μg daily using various B12-containing formulas. Treatment of the mothers is also indicated to adjust breast milk vitamin levels(Reference Green, Allen and Bjørke-Monsen260).

There is dose–response relationship between oral B12 dose and serum/plasma B12 levels(Reference Dullemeijer, Souverein and Doets687). Doubling vitamin B12 increases serum/plasma levels approximately by 11% and slightly more in the elderly (13%) than in adults (8%).

Pharmacological use

Vitamin B12 is mainly used, as mentioned above, to supplement at-risk people and to treat deficiency. In particular, older people who are more susceptible to food–cobalamin malabsorption should consume vitamin B12 in crystalline form, i.e. from supplements or fortified foods, as this form is likely to be better absorbed(Reference Allen, Miller and de Groot93).

It is speculated that vitamin B12 could be used as an adjunctive or integrative treatment for painful conditions such as various types of neuralgia and neuropathy, low back pain and aphthous stomatitis. Further studies are needed in this area(Reference Buesing, Costa, Schilling and Moeller-Bertram688,Reference Julian, Syeed and Glascow689) . Vitamin B12 could have a positive effect on sperm quality. It primarily increased sperm count and secondarily elevated sperm motility and reduced sperm DNA damage(Reference Banihani690). However, in both cases, further studies are needed as well.

Toxicity

To date, there are minimal claims on cobalamin toxicity. Vitamin B12 is usually well tolerated with rare incidence of adverse effects. Allergic reactions are unusual but may be anaphylactic(Reference Caballero, Lukawska, Lee and Dugué691,Reference Tordjman, Genereau and Guinnepain692) . Injectable forms appear to be more allergenic than pills, but this could be associated rather with some preservatives (e.g. benzylalcohol) in injections than by the vitamin itself(Reference Wolffenbuttel, Owen, Ward and Green2). High oral dose supplementation seems to be better tolerated, but there are known exceptions(Reference James and Warin693). Although there is a cross-reaction between hydroxocobalamin and cyanocobalamin, it is possible to reintroduce vitamin B12 with concomitant administration of glucocorticoids or antihistamines or with desensitisation therapy(Reference Caballero, Lukawska, Lee and Dugué691,Reference Tordjman, Genereau and Guinnepain692) . There are also cases of acneiform eruptions after intramuscular or high oral doses of vitamin B12, which easily disappear after discontinuation(Reference Morales-Gutierrez, Díaz-Cortés, Montoya-Giraldo and Zuluaga694,Reference Veraldi, Benardon, Diani and Barbareschi695) . Nausea, dry mouth and blurred vision were reported after administration of vitamin B12 as well(Reference Campbell, Heydarian and Ochoa696). Other rare documented side effects of vitamin B12 include discolouration of the skin and urine, mild arterial hypertension, hypokalaemia, congestive heart failure, pulmonary oedema and local pain at the injection site in case of parenteral administration(Reference Manzanares and Hardy578).

Conclusion

This review summarised all critical aspects of vitamin B12 biology. It emphasised that animal-based diets are almost the sole source of this vitamin for humans and, hence, vegetarians are at risk of developing its deficiency, which can progress to very severe and irreversible stages. At present, many countries have mandatory or voluntary fortification of food by this vitamin, but the risk of vitamin B12 deficiency is still not negligible worldwide. Moreover, there are many diseases, drugs and surgical procedures which can cause its deficiency, which is mostly delayed by several years as humans have relatively large stores of this vitamin. Prevention or treatment should be ideally case specific due to the complicated absorption mechanism of this vitamin. There are no convincing data on the pharmacological administration of vitamin B12 with exception of its deficiency. Its administration is considered safe in the majority of patients, but hypersensitive reactions can occur particularly after its parenteral administration.

Financial support

This open-access review paper was supported by the Erasmus+ Programme of the European Union, Key Action 2: Strategic Partnerships, project no. 2020-1-CZ01-KA203-078218. M.M. thanks the Charles University (SVV 260 663). L.K.K. thanks MH-CZ-DRO (UHHK, 00179906).

Competing interests

None.

Authorship

M.M. wrote the initial draft of the whole paper. T.S. prepared the parts related to sources of the vitamin, while L.K.K. and K.M. prepared that in relation to the detection of the vitamin. P.M. prepared the concept and revised the prepared paper. All authors revised the paper before submission and as a part of the peer-review process.

References

Hrubša, M, Siatka, T, Nejmanová, I, Vopršalová, M, Kujovská Krčmová, L, Matoušová, K, et al. (2022) Biological properties of vitamins of the B-complex, part 1: vitamins B1, B2, B3, and B5. Nutrients 14, 484.CrossRefGoogle ScholarPubMed
Wolffenbuttel, BH, Owen, PJ, Ward, M, Green, R (2023) Vitamin B(12). BMJ 383, e071725.CrossRefGoogle ScholarPubMed
Flippo, TS, Holder, WD Jr. (1993) Neurologic degeneration associated with nitrous oxide anesthesia in patients with vitamin B12 deficiency. Arch Surg 128, 13911395.CrossRefGoogle ScholarPubMed
U.S. Centers for Disease Control and Prevention (2012) Second National Report on Biochemical Indicators of Diet and Nutrition in the U.S. Population 2012, Atlanta (GA): National Center for Environmental Health.Google Scholar
O’Leary, F, Samman, S (2010) Vitamin B12 in health and disease. Nutrients 2, 299316.CrossRefGoogle ScholarPubMed
Smith, AD, Warren, MJ, Refsum, H (2018) Chapter six - vitamin B12. In Adv Food Nutr Res 83, 215279.CrossRefGoogle Scholar
Martens, JH, Barg, H, Warren, MJ, Jahn, D (2002) Microbial production of vitamin B12. Appl Microbiol Biotechnol 58, 275285.CrossRefGoogle ScholarPubMed
Yilmaz, LS, Walhout, AJ (2014) Worms, bacteria, and micronutrients: an elegant model of our diet. Trends Genet 30, 496503.CrossRefGoogle ScholarPubMed
Warren, MJ, Raux, E, Schubert, HL, Escalante-Semerena, JC (2002) The biosynthesis of adenosylcobalamin (vitamin B12). Nat Prod Rep 19, 390412.CrossRefGoogle ScholarPubMed
Grant, MA, Kazamia, E, Cicuta, P, Smith, AG (2014) Direct exchange of vitamin B12 is demonstrated by modelling the growth dynamics of algal–bacterial cocultures. ISME J 8, 14181427.CrossRefGoogle ScholarPubMed
Croft, MT, Lawrence, AD, Raux-Deery, E et al. (2005) Algae acquire vitamin B12 through a symbiotic relationship with bacteria. Nature 438, 9093.CrossRefGoogle ScholarPubMed
Tang, YZ, Koch, F, Gobler, CJ (2010) Most harmful algal bloom species are vitamin B1 and B12 auxotrophs. Proc Natl Acad Sci 107, 2075620761.CrossRefGoogle ScholarPubMed
Helliwell, KE, Lawrence, AD, Holzer, A et al. (2016) Cyanobacteria and eukaryotic algae use different chemical variants of vitamin B12. Curr Biol 26, 9991008.CrossRefGoogle ScholarPubMed
Bito, T, Teng, F, Ohishi, N et al. (2014) Characterization of vitamin B12 compounds in the fruiting bodies of shiitake mushroom (Lentinula edodes) and bed logs after fruiting of the mushroom. Mycoscience 55, 462468.CrossRefGoogle Scholar
Amin, SA, Parker, MS, Armbrust, EV (2012) Interactions between diatoms and bacteria. Microbiol Mol Biol Rev 76, 667684.CrossRefGoogle ScholarPubMed
Tandon, P, Jin, Q, Huang, L (2017) A promising approach to enhance microalgae productivity by exogenous supply of vitamins. Microb Cell Fact 16, 113.CrossRefGoogle ScholarPubMed
Sanudo-Wilhelmy, SA, Gomez-Consarnau, L, Suffridge, C, Webb, EA (2014) The role of B vitamins in marine biogeochemistry. Ann Rev Mar Sci 6, 339367.CrossRefGoogle ScholarPubMed
Roth, JR, Lawrence, J, Bobik, T (1996) Cobalamin (coenzyme B12): synthesis and biological significance. Annu Rev Microbiol 50, 137181.CrossRefGoogle ScholarPubMed
Ma, AT, Beld, J, Brahamsha, B (2017) An amoebal grazer of cyanobacteria requires cobalamin produced by heterotrophic bacteria. Appl Environ Microbiol 83, e0003500017.CrossRefGoogle ScholarPubMed
Smith, AG, Croft, MT, Moulin, M, Webb, ME (2007) Plants need their vitamins too. Curr Opin Plant Biol 10, 266275.CrossRefGoogle ScholarPubMed
Nakos, M, Pepelanova, I, Beutel, S et al. (2017) Isolation and analysis of vitamin B12 from plant samples. Food Chem 216, 301308.CrossRefGoogle Scholar
Orłowska, M, Steczkiewicz, K, Muszewska, A (2021) Utilization of cobalamin is ubiquitous in early-branching fungal phyla. Genome Biol Evol 13, evab043.CrossRefGoogle ScholarPubMed
Kennedy, KJ, Taga, ME (2020) Cobamides. Curr Biol 30, R55R56.CrossRefGoogle ScholarPubMed
Heal, KR, Qin, W, Ribalet, F et al. (2017) Two distinct pools of B12 analogs reveal community interdependencies in the ocean. Proc Natl Acad Sci 114, 364369.CrossRefGoogle ScholarPubMed
Rodionov, DA, Vitreschak, AG, Mironov, AA, Gelfand, MS (2003) Comparative genomics of the vitamin B12 metabolism and regulation in prokaryotes. J Biol Chem 278, 4114841159.CrossRefGoogle ScholarPubMed
Bertrand, EM, Saito, MA, Jeon, YJ, Neilan, BA (2011) Vitamin B12 biosynthesis gene diversity in the Ross Sea: the identification of a new group of putative polar B12 biosynthesizers. Environ Microbiol 13, 12851298.CrossRefGoogle ScholarPubMed
Lu, X, Heal, KR, Ingalls, AE et al. (2020) Metagenomic and chemical characterization of soil cobalamin production. ISME J 14, 5366.CrossRefGoogle ScholarPubMed
Shelton, AN, Seth, EC, Mok, KC et al. (2019) Uneven distribution of cobamide biosynthesis and dependence in bacteria predicted by comparative genomics. ISME J 13, 789804.CrossRefGoogle ScholarPubMed
Crofts Terence, S, Seth Erica, C, Hazra Amrita, B, Taga Michiko, E (2013) Cobamide structure depends on both lower ligand availability and CobT substrate specificity. Chem Biol 20, 12651274.CrossRefGoogle ScholarPubMed
Sokolovskaya, OM, Shelton, AN, Taga, ME (2020) Sharing vitamins: cobamides unveil microbial interactions. Science 369, eaba0165.CrossRefGoogle ScholarPubMed
Magnusdottir, S, Ravcheev, D, de Crecy-Lagard, V, Thiele, I (2015) Systematic genome assessment of B-vitamin biosynthesis suggests co-operation among gut microbes. Front Genet 6, 148.CrossRefGoogle ScholarPubMed
Zhou, J, Yu, X, Liu, J et al. (2021) VB12Path for accurate metagenomic profiling of microbially driven cobalamin synthesis pathways. Msystems 6, e0049700421.CrossRefGoogle ScholarPubMed
Mathur, Y, Sreyas, S, Datar, PM et al. (2020) CobT and BzaC catalyze the regiospecific activation and methylation of the 5-hydroxybenzimidazole lower ligand in anaerobic cobamide biosynthesis. J Biol Chem 295, 1052210534.CrossRefGoogle ScholarPubMed
Hazra, AB, Han, AW, Mehta, AP et al. (2015) Anaerobic biosynthesis of the lower ligand of vitamin B12. Proc Natl Acad Sci 112, 1079210797.CrossRefGoogle ScholarPubMed
Jeter, VL, Escalante-Semerena, JC (2021) Insights into the relationship between cobamide synthase and the cell membrane. mBio 12, e0021500221.CrossRefGoogle ScholarPubMed
Jeter, VL, Escalante-Semerena, JC (2022) Elevated levels of an enzyme involved in coenzyme B12 biosynthesis kills Escherichia coli . mBio 13, e0269702621.CrossRefGoogle ScholarPubMed
Bryant, DA, Hunter, CN, Warren, MJ (2020) Biosynthesis of the modified tetrapyrroles—the pigments of life. J Biol Chem 295, 68886925.CrossRefGoogle ScholarPubMed
Scott, AI (2003) Discovering nature’s diverse pathways to vitamin B12: a 35-year odyssey. J Org Chem 68, 25292539.CrossRefGoogle ScholarPubMed
Lauridsen, C, Matte, JJ, Lessard, M et al. (2021) Role of vitamins for gastro-intestinal functionality and health of pigs. Anim Feed Sci Technol 273, 114823.CrossRefGoogle Scholar
Degnan, PH, Barry, NA, Mok, KC et al. (2014) Human gut microbes use multiple transporters to distinguish vitamin B12 analogs and compete in the gut. Cell Host Microbe 15, 4757.CrossRefGoogle ScholarPubMed
Seth, EC, Taga, ME (2014) Nutrient cross-feeding in the microbial world. Front Microbiol 5, 350.CrossRefGoogle ScholarPubMed
Joglar, V, Pontiller, B, Martínez-García, S et al. (2021) Microbial plankton community structure and function responses to vitamin B12 and B1 amendments in an upwelling system. Appl Environ Microbiol 87, e0152501521.CrossRefGoogle Scholar
Putnam, EE, Goodman, AL (2020) B vitamin acquisition by gut commensal bacteria. PLoS Pathog 16, e1008208.CrossRefGoogle ScholarPubMed
Zhou, Z, Hu, R, Ni, Y et al. (2021) Genetic elucidation of quorum sensing and cobamide biosynthesis in divergent bacterial-fungal associations across the soil-mangrove root interface. Front Microbiol 12, 698385.CrossRefGoogle ScholarPubMed
Walworth, NG, Lee, MD, Suffridge, C, Qu, P, Fu, FX, Saito, MA, et al. (2018) Functional genomics and phylogenetic evidence suggest genus-wide cobalamin production by the globally distributed marine nitrogen fixer Trichodesmium . Front Microbiol 9, 189.CrossRefGoogle ScholarPubMed
Iguchi, H, Yurimoto, H, Sakai, Y (2015) Interactions of methylotrophs with plants and other heterotrophic bacteria. Microorganisms 3, 137151.CrossRefGoogle ScholarPubMed
Ivanova, E, Fedorov, D, Doronina, N, Trotsenko, YA (2006) Production of vitamin B 12 in aerobic methylotrophic bacteria. Microbiology 75, 494496.CrossRefGoogle ScholarPubMed
Balabanova, L, Averianova, L, Marchenok, M, Son, O, Tekutyeva, L (2021) Microbial and genetic resources for cobalamin (Vitamin B12) biosynthesis: from ecosystems to industrial biotechnology. Int J Mol Sci 22, 4522.CrossRefGoogle ScholarPubMed
Jiang, Q, Lin, L, Xie, F, Jin, W, Zhu, W, Wang, M, et al. (2022) Metagenomic insights into the microbe-mediated B and K2 vitamin biosynthesis in the gastrointestinal microbiome of ruminants. Microbiome 10, 116.CrossRefGoogle ScholarPubMed
Wienhausen, G, Dlugosch, L, Jarling, R, Wilkes, H, Giebel, HA, Simon, M (2022) Availability of vitamin B12 and its lower ligand intermediate α-ribazole impact prokaryotic and protist communities in oceanic systems. ISME J 16, 20022014.CrossRefGoogle ScholarPubMed
Doxey, AC, Kurtz, DA, Lynch, MD et al. (2015) Aquatic metagenomes implicate Thaumarchaeota in global cobalamin production. ISME J 9, 461471.CrossRefGoogle ScholarPubMed
Osman, D, Cooke, A, Young, TR et al. (2021) The requirement for cobalt in vitamin B12: a paradigm for protein metalation. Biochim Biophys Acta Mol Cell Res 1868, 118896.CrossRefGoogle ScholarPubMed
Ramanan, R, Kim, B-H, Cho, D-H et al. (2016) Algae–bacteria interactions: evolution, ecology and emerging applications. Biotechnol Adv 34, 1429.CrossRefGoogle ScholarPubMed
Danchin, A, Braham, S (2017) Coenzyme B12 synthesis as a baseline to study metabolite contribution of animal microbiota. Microb Biotechnol 10, 688701.CrossRefGoogle ScholarPubMed
Koch, F, Hattenrath-Lehmann, TK, Goleski, JA et al. (2012) Vitamin B1 and B12 uptake and cycling by plankton communities in coastal ecosystems. Front Microbiol 3, 363.CrossRefGoogle ScholarPubMed
Kazamia, E, Czesnick, H, Nguyen, TTV et al. (2012) Mutualistic interactions between vitamin B12-dependent algae and heterotrophic bacteria exhibit regulation. Environ Microbiol 14, 14661476.CrossRefGoogle ScholarPubMed
Grossman, A (2016) Nutrient acquisition: the generation of bioactive vitamin B12 by microalgae. Curr Biol 26, R319R321.CrossRefGoogle ScholarPubMed
Bertrand, EM, Allen, AE, Dupont, CL et al. (2012) Influence of cobalamin scarcity on diatom molecular physiology and identification of a cobalamin acquisition protein. Proc Natl Acad Sci 109, E1762E1771.CrossRefGoogle ScholarPubMed
Sokolovskaya, OM, Plessl, T, Bailey, H et al. (2021) Naturally occurring cobalamin (B12) analogs can function as cofactors for human methylmalonyl-CoA mutase. Biochimie 183, 3543.CrossRefGoogle ScholarPubMed
Degnan, PH, Taga, ME, Goodman, AL (2014) Vitamin B12 as a modulator of gut microbial ecology. Cell Metab 20, 769778.CrossRefGoogle ScholarPubMed
Kysil, OA (2014) Grundlegende Untersuchungen zur mikrobiellen Synthese von Vitamin B12 in symbiotischen pflanzlichen Systemen am Beispiel der Frankia: Entwicklung neuartiger pflanzlicher Extrakte mit hohem essentiellen Vitamin B12-Gehalt.Google Scholar
Scott, JM, Molloy, AM (2012) The discovery of vitamin B12. Ann Nutr Metab 61, 239245.CrossRefGoogle Scholar
Nutrition Division of FAO/WHO (2005) Vitamin B12. In Human Vitamin and Mineral Requirements, 2nd ed. WHO, pp. 279288.Google Scholar
Basu, TK, Donaldson, D (2003) Intestinal absorption in health and disease: micronutrients. Best Pract Res Clin Gastroenterol 17, 957979.CrossRefGoogle ScholarPubMed
Herbert, V (1988) Vitamin B-12: plant sources, requirements, and assay. Am J Clin Nutr 48, 852858.CrossRefGoogle ScholarPubMed
Allen, LH (2008) Causes of vitamin B12 and folate deficiency. Food Nutr Bull 29, S20S34.CrossRefGoogle ScholarPubMed
Hohmann, HP, Litta, G, Hans, M et al. (2020) Vitamins, 13. Vitamin B12 (Cobalamins). In Ullmann’s Encyclopedia of Industrial Chemistry, 7th ed. Weinheim, Germany: Wiley-VCH Verlag GmbH & Co. KGaA, pp. 110.Google Scholar
Gille, D, Schmid, A (2015) Vitamin B12 in meat and dairy products. Nutr Rev 73, 106115.CrossRefGoogle ScholarPubMed
Biesalski, HK (2016) Nutrition meets the microbiome: micronutrients and the microbiota. Ann N Y Acad Sci 1372, 5364.CrossRefGoogle ScholarPubMed
Janice Marie Sych, CL, Stevens, Marc J.A. (2016) Vitamin B12–physiology, production and application. In Industrial biotechnology of vitamins, biopigments, and antioxidants, pp. 129-159 [JLR Erick J. Vandamme, editor].CrossRefGoogle Scholar
Rowland, I, Gibson, G, Heinken, A et al. (2018) Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr 57, 124.CrossRefGoogle ScholarPubMed
Guetterman, HM, Huey, SL, Knight, R et al. (2022) Vitamin B-12 and the gastrointestinal microbiome: a systematic review. Adv Nutr 13, 530558.CrossRefGoogle ScholarPubMed
Rowley, CA, Kendall, MM (2019) To B12 or not to B12: five questions on the role of cobalamin in host-microbial interactions. PLoS Pathog 15, e1007479.CrossRefGoogle ScholarPubMed
Allen, RH, Stabler, SP (2008) Identification and quantitation of cobalamin and cobalamin analogues in human feces. Am J Clin Nutr 87, 13241335.CrossRefGoogle ScholarPubMed
Rodionov, DA, Arzamasov, AA, Khoroshkin, MS et al. (2019) Micronutrient requirements and sharing capabilities of the human gut microbiome. Front Microbiol 10, 1316.CrossRefGoogle ScholarPubMed
Kundra, P, Geirnaert, A, Pugin, B et al. (2022) Healthy adult gut microbiota sustains its own vitamin B12 requirement in an in vitro batch fermentation model. Front Nutr 9, 1070155.CrossRefGoogle Scholar
Kurpad, AV, Pasanna, RM, Hegde, SG et al. (2023) Bioavailability and daily requirement of vitamin B12 in adult humans: an observational study of its colonic absorption and daily excretion as measured by [13C]-cyanocobalamin kinetics. Am J Clin Nutr 118, 12141223.CrossRefGoogle ScholarPubMed
Fedosov, SN (2023) New insights into mechanisms of vitamin B12 uptake and conversion. Am J Clin Nutr S0002-9165, 66163.Google ScholarPubMed
Watanabe, F, Bito, T, Koseki, K (2022) Determination of cobalamin and related compounds in foods. Vitam Horm 119, 491504.CrossRefGoogle ScholarPubMed
Watanabe, F, Bito, T (2018) Vitamin B12 sources and microbial interaction. Exp Biol Med (Maywood) 243, 148158.CrossRefGoogle ScholarPubMed
Sobczyńska-Malefora, A, Delvin, E, McCaddon, A et al. (2021) Vitamin B12 status in health and disease: a critical review. Diagnosis of deficiency and insufficiency–clinical and laboratory pitfalls. Crit Rev Clin Lab Sci 58, 399429.CrossRefGoogle Scholar
Antony, AC (2003) Vegetarianism and vitamin B-12 (cobalamin) deficiency. Am J Clin Nutr 78, 36.CrossRefGoogle ScholarPubMed
Bito, T, Tanioka, Y, Watanabe, F (2018) Characterization of vitamin B12 compounds from marine foods. Fish Sci 84, 747755.CrossRefGoogle Scholar
Watanabe, F (2007) Vitamin B12 sources and bioavailability. Exp Biol Med (Maywood) 232, 12661274.CrossRefGoogle Scholar
Watanabe, F, Yabuta, Y, Tanioka, Y, Bito, T (2013) Biologically active vitamin B12 compounds in foods for preventing deficiency among vegetarians and elderly subjects. J Agric Food Chem 61, 67696775.CrossRefGoogle ScholarPubMed
González-Montaña, J-R, Escalera-Valente, F, Alonso, AJ et al. (2020) Relationship between vitamin B12 and cobalt metabolism in domestic ruminant: an update. Animals 10, 1855.CrossRefGoogle ScholarPubMed
Franco-Lopez, J, Duplessis, M, Bui, A et al. (2020) Correlations between the composition of the bovine microbiota and vitamin B12 abundance. Msystems 5, e0010700120.CrossRefGoogle ScholarPubMed
EFSA (2015) Scientific opinion on dietary reference values for cobalamin (vitamin B12). EFSA J 13, 4150.Google Scholar
Sharma, S, Sheehy, T, Kolonel, LN (2013) Contribution of meat to vitamin B 12, iron and zinc intakes in five ethnic groups in the USA: implications for developing food-based dietary guidelines. J Hum Nutr Diet 26, 156168.CrossRefGoogle ScholarPubMed
Tucker, KL, Rich, S, Rosenberg, I et al. (2000) Plasma vitamin B-12 concentrations relate to intake source in the Framingham offspring study. Am J Clin Nutr 71, 514522.CrossRefGoogle ScholarPubMed
Vogiatzoglou, A, Smith, AD, Nurk, E et al. (2009) Dietary sources of vitamin B-12 and their association with plasma vitamin B-12 concentrations in the general population: the Hordaland Homocysteine study. Am J Clin Nutr 89, 10781087.CrossRefGoogle ScholarPubMed
Allen, LH (2010) Bioavailability of vitamin B12. Int J Vitam Nutr Res 80, 330.CrossRefGoogle ScholarPubMed
Allen, LH, Miller, JW, de Groot, L et al. (2018) Biomarkers of nutrition for development (BOND): vitamin B-12 review. J Nutr 148, 1995s2027s.CrossRefGoogle ScholarPubMed
Fedosov, SN, Nexo, E, Heegaard, CW (2019) Vitamin B12 and its binding proteins in milk from cow and buffalo in relation to bioavailability of B12. J Dairy Sci 102, 48914905.CrossRefGoogle ScholarPubMed
Yoshino, K, Inagawa, M, Oshima, M et al. (2005) Trends in dietary intake of folate, vitamins B6, and B12 among Japanese adults in two rural communities from 1974 through 2001. J Epidemiol 15, 2937.CrossRefGoogle ScholarPubMed
Duplessis, M, Fréchette, A, Poisson, W et al. (2021) Refining knowledge of factors affecting vitamin B12 concentration in bovine milk. Animals 11, 532.CrossRefGoogle ScholarPubMed
Duplessis, M, Pellerin, D, Cue, R, Girard, C (2016) Factors affecting vitamin B12 concentration in milk of commercial dairy herds: an exploratory study. J Dairy Sci 99, 48864892.CrossRefGoogle ScholarPubMed
Doets, EL, Szczecińska, A, Dhonukshe-Rutten, RA et al. (2013) Systematic review on daily vitamin B12 losses and bioavailability for deriving recommendations on vitamin B12 intake with the factorial approach. Ann Nutr Metab 62, 311322.CrossRefGoogle ScholarPubMed
Matte, J, Britten, M, Girard, C (2014) The importance of milk as a source of vitamin B12 for human nutrition. Anim Front 4, 3237.CrossRefGoogle Scholar
Matte, JJ, Guay, F, Girard, CL (2012) Bioavailability of vitamin B12 in cows’ milk. Br J Nutr 107, 6166.CrossRefGoogle ScholarPubMed
Greibe, E, Reitelseder, S, Bechshøft, RL et al. (2020) Effects of prolonged whey protein supplementation and resistance training on biomarkers of vitamin B12 status: a 1-year randomized intervention in healthy older adults (the CALM study). Nutrients 12, 2015.CrossRefGoogle ScholarPubMed
Ströhle, A, Richter, M, González-Gross, M et al. (2019) The revised D-A-CH-reference values for the intake of vitamin B12: prevention of deficiency and beyond. Mol Nutr Food Res 63, 1801178.CrossRefGoogle ScholarPubMed
Banjari, I, Hjartåker, A (2018) Dietary sources of iron and vitamin B12: is this the missing link in colorectal carcinogenesis? Med Hypotheses 116, 105110.CrossRefGoogle ScholarPubMed
Lavriša, Ž, Hristov, H, Hribar, M et al. (2022) Dietary intake and status of vitamin B12 in slovenian population. Nutrients 14, 334.CrossRefGoogle ScholarPubMed
Kumar, C, Rana, RK, Kumar, M et al. (2021) Effect of milk supplementation on the status of micronutrients among rural school children aged 5–19 years in a tribal predominating district of India. BMJ Nutr Prev Health 4, 463.CrossRefGoogle Scholar
Watanabe, F, Katsura, H, Takenaka, S et al. (2001) Characterization of vitamin B 12 compounds from edible shellfish, clam, oyster, and mussel. Int J Food Sci Nutr 52, 263268.CrossRefGoogle ScholarPubMed
Brouwer-Brolsma, EM, Dhonukshe-Rutten, RA, Van Wijngaarden, JP et al. (2015) Dietary sources of vitamin B-12 and their association with vitamin B-12 status markers in healthy older adults in the B-PROOF study. Nutrients 7, 77817797.CrossRefGoogle ScholarPubMed
Obeid, R, Heil, SG, Verhoeven, MM et al. (2019) Vitamin B12 intake from animal foods, biomarkers, and health aspects. Front Nutr 6, 93.CrossRefGoogle ScholarPubMed
Górska-Warsewicz, H, Rejman, K, Laskowski, W, Czeczotko, M (2019) Milk and dairy products and their nutritional contribution to the average polish diet. Nutrients 11, 1771.CrossRefGoogle Scholar
Auclair, O, Han, Y, Burgos, SA (2019) Consumption of milk and alternatives and their contribution to nutrient intakes among Canadian adults: evidence from the 2015 Canadian Community Health Survey-Nutrition. Nutrients 11, 1948.CrossRefGoogle ScholarPubMed
Li, C (2017) The role of beef in human nutrition and health. In Ensuring safety and quality in the production of beef, vol. 2, pp. 329-338 [ME Dikeman, editor].CrossRefGoogle Scholar
Devi, A, Rush, E, Harper, M, Venn, B (2018) Vitamin B12 status of various ethnic groups living in New Zealand: an analysis of the adult nutrition survey 2008/2009. Nutrients 10, 181.CrossRefGoogle ScholarPubMed
Denissen, KF, Heil, SG, Eussen, SJ et al. (2019) Intakes of vitamin B-12 from dairy food, meat, and fish and shellfish are independently and positively associated with vitamin b-12 biomarker status in pregnant dutch women. J Nutr 149, 131138.CrossRefGoogle ScholarPubMed
Kwak, CS, Lee, MS, Oh, SI, Park, SC (2010) Discovery of novel sources of vitamin B12 in traditional Korean foods from nutritional surveys of centenarians. Curr Gerontol Geriatr Res 2010, 374897.CrossRefGoogle ScholarPubMed
Kwak, CS, Lee, MS, Lee, HJ et al. (2010) Dietary source of vitamin B12 intake and vitamin B12 status in female elderly Koreans aged 85 and older living in rural area. Nutr Res Pract 4, 229234.CrossRefGoogle ScholarPubMed
Wyness, L (2016) The role of red meat in the diet: nutrition and health benefits. Proc Nutr Soc 75, 227232.CrossRefGoogle ScholarPubMed
Wyness, L, Weichselbaum, E, O’connor, A et al. (2011) Red meat in the diet: an update. Nutr Bull 36, 3477.CrossRefGoogle Scholar
Pereira, PMDCC, Vicente, AFDRB (2013) Meat nutritional composition and nutritive role in the human diet. Meat Sci 93, 586592.CrossRefGoogle ScholarPubMed
Cosgrove, M, Flynn, A, Kiely, M (2005) Consumption of red meat, white meat and processed meat in Irish adults in relation to dietary quality. Br J Nutr 93, 933942.CrossRefGoogle ScholarPubMed
Muehlhoff, E, Bennett, A, McMahon, D (2013) Milk and dairy products in human nutrition. Rome, Italy: FAO.Google Scholar
Zironi, E, Gazzotti, T, Barbarossa, A et al. (2014) Determination of vitamin B12 in dairy products by ultra performance liquid chromatography-tandem mass spectrometry. Ital J Food Saf 3.Google ScholarPubMed
Shetty, SA, Young, MF, Taneja, S, Rangiah, K (2020) Quantification of B-vitamins from different fresh milk samples using ultra-high performance liquid chromatography mass spectrometry/selected reaction monitoring methods. J Chromatogr A 1609, 460452.CrossRefGoogle ScholarPubMed
Guggisberg, D, Risse, M, Hadorn, R (2012) Determination of vitamin B12 in meat products by RP-HPLC after enrichment and purification on an immunoaffinity column. Meat Sci 90, 279283.CrossRefGoogle Scholar
Li, Y, Gill, BD, Grainger, MN, Manley-Harris, M (2019) The analysis of vitamin B12 in milk and infant formula: a review. Int Dairy J 99, 104543.CrossRefGoogle Scholar
Pérez-Fernández, V, Gentili, A, Martinelli, A et al. (2016) Evaluation of oxidized buckypaper as material for the solid phase extraction of cobalamins from milk: its efficacy as individual and support sorbent of a hydrophilic–lipophilic balance copolymer. J Chromatogr A 1428, 255266.CrossRefGoogle ScholarPubMed
Esteve, M, Farré, R, Frıgola, A, Pilamunga, C (2002) Contents of vitamins B1, B2, B6, and B12 in pork and meat products. Meat Sci 62, 7378.CrossRefGoogle ScholarPubMed
Iglesia, I, Mouratidou, T, Gonzalez-Gross, M et al. (2017) Foods contributing to vitamin B 6, folate, and vitamin B 12 intakes and biomarkers status in European adolescents: the HELENA study. Eur J Nutr 56, 17671782.CrossRefGoogle ScholarPubMed
Geiker, NRW, Bertram, HC, Mejborn, H et al. (2021) Meat and human health—current knowledge and research gaps. Foods 10, 1556.CrossRefGoogle ScholarPubMed
Kraemer, K, Semba, RD, Eggersdorfer, M, Schaumberg, DA (2012) Introduction: the diverse and essential biological functions of vitamins. Ann Nutr Metab 61, 185.CrossRefGoogle ScholarPubMed
Williams, P (2007) Nutritional composition of red meat. Nutr Diet 64, S113S119.CrossRefGoogle Scholar
Arkbåge, K (2003) Vitamin B12, folate and folate-binding proteins in dairy products. vol. 430.Google Scholar
Rampazzo, G, Zironi, E, Pagliuca, G, Gazzotti, T (2022) Analysis of cobalamin (Vit B12) in ripened cheese by ultra-high-performance liquid chromatography coupled with Mass spectrometry. Foods 11, 2745.CrossRefGoogle ScholarPubMed
Melse-Boonstra, A (2020) Bioavailability of micronutrients from nutrient-dense whole foods: zooming in on dairy, vegetables, and fruits. Front Nutr 7, 101.CrossRefGoogle Scholar
Bodwell, C, Anderson, B (1986) Nutritional composition and value of meat and meat products, Muscle as food. Orlando, USA: Academic Press.Google Scholar
Partearroyo, T, Samaniego-Vaesken, MDL, Ruiz, E et al. (2017) Dietary sources and intakes of folates and vitamin B12 in the Spanish population: findings from the ANIBES study. PLoS One 12, e0189230.CrossRefGoogle ScholarPubMed
Planells, E, Sanchez, C, Montellano, M et al. (2003) Vitamins B6 and B12 and folate status in an adult Mediterranean population. Eur J Clin Nutr 57, 777785.CrossRefGoogle Scholar
Truswell, AS (2007) Vitamin B12. Nutr Diet 64, S120S120.CrossRefGoogle Scholar
Ahmad, RS, Imran, A, Hussain, MB (2018) Nutritional composition of meat. In Meat science and nutrition [Arshad, MS, editor]. London, UK: IntechOpen Limited.Google Scholar
Xu, J, Clare, CE, Brassington, AH et al. (2020) Comprehensive and quantitative profiling of B vitamins and related compounds in the mammalian liver. J Chromatogr B 1136, 121884.CrossRefGoogle ScholarPubMed
Gherghina, E, Israel-Roming, F, Balan, D et al. (2021) Assessment of vitamin content in different types of Romanian cheese. Rom Biotechnol Lett 26, 23752383.CrossRefGoogle Scholar
Azzini, E, Raguzzini, A, Polito, A (2021) A brief review on vitamin B12 deficiency looking at some case study reports in adults. Int J Mol Sci 22, 9694.CrossRefGoogle ScholarPubMed
Watanabe, F, Bito, T (2016) Corrinoids in food and biological samples. Front Nat Prod Chem 2, 229244.CrossRefGoogle Scholar
Watanabe, F, Bito, T (2018) Determination of cobalamin and related compounds in foods. J AOAC Int 101, 13081313.CrossRefGoogle ScholarPubMed
Kwak, CS, Park, JH, Cho, JH (2012) Vitamin B12 content using modified microbioassay in some Korean popular seaweeds, fish, shellfish and its products. Korean J Nutr 45, 94102.CrossRefGoogle Scholar
Okamoto, N, Hamaguchi, N, Umebayashi, Y et al. (2020) Determination and characterization of vitamin B 12 in the muscles and head innards of edible shrimp. Fish Sci 86, 395406.CrossRefGoogle Scholar
Teng, F, Tanioka, Y, Hamaguchi, N et al. (2015) Determination and characterization of vitamin B 12 compounds in edible sea snails, ivory shell Babylonia japonica and turban shell Turdo Batillus cornutus . Fish Sci 81, 11051111.CrossRefGoogle Scholar
Tanioka, Y, Takenaka, S, Furusho, T et al. (2014) Identification of vitamin B 12 and pseudovitamin B 12 from various edible shellfish using liquid chromatography–electrospray ionization/tandem mass spectrometry. Fish Sci 80, 10651071.CrossRefGoogle Scholar
Stupperich, E, NEXØ, E (1991) Effect of the cobalt-N coordination on the cobamide recognition by the human vitamin B12 binding proteins intrinsic factor, transcobalamin and haptocorrin. Eur J Biochem 199, 299303.CrossRefGoogle ScholarPubMed
Fedosov, SN, Fedosova, NU, Kräutler, B et al. (2007) Mechanisms of discrimination between cobalamins and their natural analogues during their binding to the specific B12-transporting proteins. Biochemistry 46, 64466458.CrossRefGoogle Scholar
Bito, T, Teng, F, Watanabe, F (2017) Bioactive compounds of edible purple laver Porphyra sp. (Nori). J Agric Food Chem 65, 1068510692.CrossRefGoogle ScholarPubMed
Bito, T, Bito, M, Hirooka, T et al. (2020) Biological activity of pseudovitamin B12 on cobalamin-dependent methylmalonyl-CoA mutase and methionine synthase in mammalian cultured COS-7 cells. Molecules 25, 3268.CrossRefGoogle ScholarPubMed
Stabler, SP (2020) Vitamin B12. In Present knowledge in nutrition, 11th ed., pp. 257271 [Bernadette, DFB, Marriott, P., … Yates, Allison A., editor] Elsevier.CrossRefGoogle Scholar
Schmidt, A, Call, L-M, Macheiner, L, Mayer, HK (2019) Determination of vitamin B12 in four edible insect species by immunoaffinity and ultra-high performance liquid chromatography. Food Chem 281, 124129.CrossRefGoogle ScholarPubMed
Nowak, V, Persijn, D, Rittenschober, D, Charrondiere, UR (2016) Review of food composition data for edible insects. Food Chem 193, 3946.CrossRefGoogle ScholarPubMed
Okamoto, N, Nagao, F, Umebayashi, Y et al. (2021) Pseudovitamin B12 and factor S are the predominant corrinoid compounds in edible cricket products. Food Chem 347, 129048.CrossRefGoogle Scholar
Bawa, M, Songsermpong, S, Kaewtapee, C, Chanput, W (2020) Effect of diet on the growth performance, feed conversion, and nutrient content of the house cricket. J Insect Sci 20, 10.CrossRefGoogle ScholarPubMed
Watanabe, F, Schwarz, J, Takenaka, S et al. (2012) Characterization of vitamin B12 compounds in the wild edible mushrooms black trumpet (Craterellus cornucopioides) and golden chanterelle (Cantharellus cibarius). J Nutr Sci Vitaminol 58, 438441.CrossRefGoogle ScholarPubMed
de Brito, M, Campos, B, Menezes, D et al. (2022) Vitamin B12 sources in non-animal foods: a systematic review. Crit Rev Food Sci Nutr, 115.Google Scholar
Koyyalamudi, SR, Jeong, S-C, Cho, KY, Pang, G (2009) Vitamin B12 is the active corrinoid produced in cultivated white button mushrooms (Agaricus bisporus). J Agric Food Chem 57, 63276333.CrossRefGoogle ScholarPubMed
Teng, F, Bito, T, Takenaka, S et al. (2014) Vitamin B12 [c-lactone], a biologically inactive corrinoid compound, occurs in cultured and dried lion’s mane mushroom (Hericium erinaceus) fruiting bodies. J Agric Food Chem 62, 17261732.CrossRefGoogle ScholarPubMed
Teng, F, Bito, T, Takenaka, S et al. (2015) Determination and characterization of corrinoid compounds in truffle (Tuber spp.) and shoro (Rhizopogon rubescens) fruiting bodies. Mushroom Sci Biotechnol 22, 159164.Google Scholar
La Guardia, M, Venturella, G, Venturella, F (2005) On the chemical composition and nutritional value of Pleurotus taxa growing on umbelliferous plants (Apiaceae). J Agric Food Chem 53, 59976002.CrossRefGoogle ScholarPubMed
Rizzo, G, Laganà, AS, Rapisarda, AMC et al. (2016) Vitamin B12 among vegetarians: status, assessment and supplementation. Nutrients 8, 767.CrossRefGoogle ScholarPubMed
Mattila, P, Konko, K, Eurola, M et al. (2001) Contents of vitamins, mineral elements, and some phenolic compounds in cultivated mushrooms. J Agric Food Chem 49, 23432348.CrossRefGoogle ScholarPubMed
Juszczyk, P, Smolczyk, A, Gil, Z et al. (2015) Produkcja drożdży paszowych Yarrowia lipolytica wzbogaconych w aminokwasy selenowe i witaminę B12. Inżynieria i Aparatura Chemiczna.Google Scholar
Jach, ME, Masłyk, M, Juda, M et al. (2020) Vitamin B12-enriched Yarrowia lipolytica biomass obtained from biofuel waste. Waste Biomass Valorization 11, 17111716.CrossRefGoogle Scholar
Jach, ME, Malm, A (2022) Yarrowia lipolytica as an alternative and valuable source of nutritional and bioactive compounds for humans. Molecules 27, 2300.CrossRefGoogle ScholarPubMed
Brawley, SH, Blouin, NA, Ficko-Blean, E et al. (2017) Insights into the red algae and eukaryotic evolution from the genome of Porphyra umbilicalis (Bangiophyceae, Rhodophyta). Proc Natl Acad Sci 114, E6361E6370.CrossRefGoogle ScholarPubMed
Helliwell, KE (2017) The roles of B vitamins in phytoplankton nutrition: new perspectives and prospects. New Phytol 216, 6268.CrossRefGoogle ScholarPubMed
Helliwell, KE, Pandhal, J, Cooper, MB et al. (2018) Quantitative proteomics of a B12-dependent alga grown in coculture with bacteria reveals metabolic tradeoffs required for mutualism. New Phytol 217, 599612.CrossRefGoogle Scholar
van den Oever, SP, Mayer, HK (2022) Biologically active or just “pseudo”-vitamin B12 as predominant form in algae-based nutritional supplements? J Food Compost Anal 109, 104464.CrossRefGoogle Scholar
Bito, T, Bito, M, Asai, Y et al. (2016) Characterization and quantitation of vitamin B12 compounds in various chlorella supplements. J Agric Food Chem 64, 85168524.CrossRefGoogle Scholar
Nef, C, Jung, S, Mairet, F et al. (2019) How haptophytes microalgae mitigate vitamin B12 limitation. Sci Rep 9, 8417.CrossRefGoogle ScholarPubMed
Nef, C, Dittami, S, Kaas, R et al. (2022) Sharing vitamin B12 between bacteria and microalgae does not systematically occur: case study of the haptophyte Tisochrysis lutea . Microorganisms 10, 1337.CrossRefGoogle Scholar
Pereira, J, Simões, M, Silva, JL (2019) Microalgal assimilation of vitamin B12 toward the production of a superfood. J Food Biochem 43, e12911.CrossRefGoogle ScholarPubMed
Kanazawa, A (1963) Vitamins in algae. Nippon Suisan Gakkai Shi 29, 713731.CrossRefGoogle Scholar
Smith, EM, Hoi, JT, Eissenberg, JC et al. (2007) Feeding Drosophila a biotin-deficient diet for multiple generations increases stress resistance and lifespan and alters gene expression and histone biotinylation patterns. J Nutr 137, 20062012.CrossRefGoogle Scholar
Croft, MT, Warren, MJ, Smith, AG (2006) Algae need their vitamins. Eukaryot Cell 5, 11751183.CrossRefGoogle ScholarPubMed
Kazamia, E, Helliwell, KE, Purton, S, Smith, AG (2016) How mutualisms arise in phytoplankton communities: building eco-evolutionary principles for aquatic microbes. Ecol Lett 19, 810822.CrossRefGoogle ScholarPubMed
Wells, ML, Potin, P, Craigie, JS et al. (2017) Algae as nutritional and functional food sources: revisiting our understanding. J Appl Phycol 29, 949982.CrossRefGoogle ScholarPubMed
Watanabe, F, Takenaka, S, Katsura, H et al. (1999) Dried green and purple lavers (Nori) contain substantial amounts of biologically active vitamin B12 but less of dietary iodine relative to other edible seaweeds. J Agric Food Chem 47, 23412343.CrossRefGoogle ScholarPubMed
Watanabe, F, Takenaka, S, Kittaka-Katsura, H et al. (2002) Characterization and bioavailability of vitamin B12-compounds from edible algae. J Nutr Sci Vitaminol 48, 325331.CrossRefGoogle ScholarPubMed
Watanabe, F, Takenaka, S, Katsura, H et al. (2000) Characterization of a vitamin B12 compound in the edible purple laver, Porphyra yezoensis . Biosci Biotechnol Biochem 64, 27122715.CrossRefGoogle ScholarPubMed
Watanabe, F, Yabuta, Y, Bito, T, Teng, F (2014) Vitamin B12-containing plant food sources for vegetarians. Nutrients 6, 18611873.CrossRefGoogle ScholarPubMed
Bito, T, Watanabe, F (2022) Seaweeds as a Source of Vitamin B12. In Sustainable Global Resources of Seaweeds Volume 2: Food, Pharmaceutical and Health Applications, pp. 339 [GAR Ambati Ranga Rao, editor].CrossRefGoogle Scholar
Takenaka, S, Sugiyama, S, Ebara, S et al. (2001) Feeding dried purple laver (nori) to vitamin B12-deficient rats significantly improves vitamin B12 status. Br J Nutr 85, 699703.CrossRefGoogle ScholarPubMed
Miyamoto, E, Yabuta, Y, Kwak, CS et al. (2009) Characterization of vitamin B12 compounds from Korean purple laver (Porphyra sp.) products. J Agric Food Chem 57, 27932796.CrossRefGoogle ScholarPubMed
Martínez–Hernández, GB, Castillejo, N, Carrión–Monteagudo, MDM et al. (2018) Nutritional and bioactive compounds of commercialized algae powders used as food supplements. Food Sci Technol Int 24, 172182.CrossRefGoogle ScholarPubMed
Kittaka-Katsura, H, Fujita, T, Watanabe, F, Nakano, Y (2002) Purification and characterization of a corrinoid compound from Chlorella tablets as an algal health food. J Agric Food Chem 50, 49944997.CrossRefGoogle ScholarPubMed
Yagame, BM, Mensah, ANC, Mady, C et al. (2017) Nutritional composition of Meristotheca senegalense (Rhodophyta): a new nutrient source. Afr J Food Sci 11, 1217.Google Scholar
Edelmann, M, Aalto, S, Chamlagain, B et al. (2019) Riboflavin, niacin, folate and vitamin B12 in commercial microalgae powders. J Food Compost Anal 82, 103226.CrossRefGoogle Scholar
Merchant, RE, Phillips, TW, Udani, J (2015) Nutritional supplementation with Chlorella pyrenoidosa lowers serum methylmalonic acid in vegans and vegetarians with a suspected vitamin B12 deficiency. J Med Food 18, 13571362.CrossRefGoogle ScholarPubMed
Cherry, P, O’Hara, C, Magee, PJ et al. (2019) Risks and benefits of consuming edible seaweeds. Nutr Rev 77, 307329.CrossRefGoogle ScholarPubMed
Kaplan, A, Zelicha, H, Tsaban, G et al. (2019) Protein bioavailability of Wolffia globosa duckweed, a novel aquatic plant – a randomized controlled trial. Clin Nutr 38, 25762582.CrossRefGoogle ScholarPubMed
Sela, I, Yaskolka Meir, A, Brandis, A et al. (2020) Wolffia globosa–mankai plant-based protein contains bioactive vitamin B12 and is well absorbed in humans. Nutrients 12, 3067.CrossRefGoogle ScholarPubMed
Kittaka-Katsura, H, Watanabe, F, Nakano, Y (2004) Occurrence of vitamin B12 in green, blue, red, and black tea leaves. J Nutr Sci Vitaminol 50, 438440.CrossRefGoogle Scholar
Lawrence, AD, Nemoto-Smith, E, Deery, E et al. (2018) Construction of fluorescent analogs to follow the uptake and distribution of cobalamin (vitamin B12) in bacteria, worms, and plants. Cell Chem Biol 25, 941951. e946.CrossRefGoogle ScholarPubMed
Ofoedu, CE, Iwouno, JO, Ofoedu, EO et al. (2021) Revisiting food-sourced vitamins for consumer diet and health needs: a perspective review, from vitamin classification, metabolic functions, absorption, utilization, to balancing nutritional requirements. PeerJ 9, e11940.CrossRefGoogle Scholar
Tucker, KL, Olson, B, Bakun, P et al. (2004) Breakfast cereal fortified with folic acid, vitamin B-6, and vitamin B-12 increases vitamin concentrations and reduces homocysteine concentrations: a randomized trial. Am J Clin Nutr 79, 805811.CrossRefGoogle ScholarPubMed
Winkels, RM, Brouwer, IA, Clarke, R et al. (2008) Bread cofortified with folic acid and vitamin B-12 improves the folate and vitamin B-12 status of healthy older people: a randomized controlled trial. Am J Clin Nutr 88, 348355.CrossRefGoogle ScholarPubMed
Naik, S, Mahalle, N, Greibe, E et al. (2019) Cyano-B12 or whey powder with endogenous hydroxo-B12 for supplementation in B12 deficient lactovegetarians. Nutrients 11, 2382.CrossRefGoogle ScholarPubMed
Yajnik, C, Deshpande, SS, Lubree, HG et al. (2006) Vitamin B12 deficiency and hyperhomocysteinemia in rural and urban Indians. JAPI 54, 82.Google ScholarPubMed
Allen, LH (2008) To what extent can food-based approaches improve micronutrient status? Asia Pac J Clin Nutr 17, 103105.Google ScholarPubMed
Craig, WJ (2009) Health effects of vegan diets. Am J Clin Nutr 89, 1627S1633S.CrossRefGoogle ScholarPubMed
Melina, V, Craig, W, Levin, S (2016) Position of the academy of nutrition and dietetics: vegetarian diets. J Acad Nutr Diet 116, 19701980.CrossRefGoogle ScholarPubMed
McEvoy, CT, Temple, N, Woodside, JV (2012) Vegetarian diets, low-meat diets and health: a review. Public Health Nutr 15, 22872294.CrossRefGoogle ScholarPubMed
Hargreaves, SM, Raposo, A, Saraiva, A, Zandonadi, RP (2021) Vegetarian diet: an overview through the perspective of quality of life domains. Int J Environ Res Public Health 18, 4067.CrossRefGoogle ScholarPubMed
Pellinen, T, Päivärinta, E, Isotalo, J et al. (2022) Replacing dietary animal-source proteins with plant-source proteins changes dietary intake and status of vitamins and minerals in healthy adults: a 12-week randomized controlled trial. Eur J Nutr, 114.Google ScholarPubMed
Gilsing, AM, Crowe, FL, Lloyd-Wright, Z et al. (2010) Serum concentrations of vitamin B12 and folate in British male omnivores, vegetarians and vegans: results from a cross-sectional analysis of the EPIC-Oxford cohort study. Eur J Clin Nutr 64, 933939.CrossRefGoogle ScholarPubMed
Lindqvist, HM, Rådjursöga, M, Malmodin, D et al. (2019) Serum metabolite profiles of habitual diet: evaluation by 1H-nuclear magnetic resonance analysis. Am J Clin Nutr 110, 5362.CrossRefGoogle ScholarPubMed
Obersby, D, Chappell, DC, Dunnett, A, Tsiami, AA (2013) Plasma total homocysteine status of vegetarians compared with omnivores: a systematic review and meta-analysis. Br J Nutr 109, 785794.CrossRefGoogle ScholarPubMed
Malik, A, Trilok-Kumar, G (2020) Status of vitamin B12 among healthy adult and elderly population in India: a review. J Nutr Sci Vitaminol 66, S361S368.CrossRefGoogle ScholarPubMed
Sobiecki, JG, Appleby, PN, Bradbury, KE, Key, TJ (2016) High compliance with dietary recommendations in a cohort of meat eaters, fish eaters, vegetarians, and vegans: results from the European Prospective Investigation into Cancer and Nutrition–Oxford study. Nutr Res 36, 464477.CrossRefGoogle Scholar
Stabler, SP, Allen, RH (2004) Vitamin B12 deficiency as a worldwide problem. Annu Rev Nutr 24, 299326.CrossRefGoogle ScholarPubMed
Naik, S, Mahalle, N, Bhide, V (2018) Identification of vitamin B12 deficiency in vegetarian Indians. Br J Nutr 119, 629635.CrossRefGoogle ScholarPubMed
Sebastiani, G, Herranz Barbero, A, Borrás-Novell, C et al. (2019) The effects of vegetarian and vegan diet during pregnancy on the health of mothers and offspring. Nutrients 11, 557.CrossRefGoogle ScholarPubMed
Shaw, KA, Zello, GA, Rodgers, CD et al. (2022) Benefits of a plant-based diet and considerations for the athlete. Eur J Appl Physiol 122, 11631178.CrossRefGoogle ScholarPubMed
Rogerson, D (2017) Vegan diets: practical advice for athletes and exercisers. J Int Soc Sports Nutr 14, 36.CrossRefGoogle ScholarPubMed
Haddad, EH, Jaceldo-Siegl, K, Oda, K, Fraser, GE (2020) Associations of circulating methylmalonic acid and vitamin B-12 biomarkers are modified by vegan dietary pattern in adult and elderly participants of the adventist health study 2 calibration study. Curr Dev Nutr 4, nzaa008.CrossRefGoogle ScholarPubMed
Allès, B, Baudry, J, Méjean, C et al. (2017) Comparison of sociodemographic and nutritional characteristics between self-reported vegetarians, vegans, and meat-eaters from the NutriNet-Santé study. Nutrients 9, 1023.CrossRefGoogle ScholarPubMed
Světnička, M, Sigal, A, Selinger, E et al. (2022) Cross-sectional study of the prevalence of cobalamin deficiency and vitamin B12 supplementation habits among vegetarian and vegan children in the Czech Republic. Nutrients 14, 535.CrossRefGoogle ScholarPubMed
Phillips, F (2005) Vegetarian nutrition. Nutr Bull 30, 132167.CrossRefGoogle Scholar
Herrmann, W, Schorr, H, Obeid, R, Geisel, J (2003) Vitamin B-12 status, particularly holotranscobalamin II and methylmalonic acid concentrations, and hyperhomocysteinemia in vegetarians. Am J Clin Nutr 78, 131136.CrossRefGoogle ScholarPubMed
Zeuschner, CL, Hokin, BD, Marsh, KA et al. (2013) Vitamin B12 and vegetarian diets. Med J Aust 199, S27S32.CrossRefGoogle ScholarPubMed
Elorinne, A-L, Alfthan, G, Erlund, I et al. (2016) Food and nutrient intake and nutritional status of Finnish vegans and non-vegetarians. PLoS One 11, e0148235.CrossRefGoogle Scholar
Pawlak, R, Parrott, SJ, Raj, S et al. (2013) How prevalent is vitamin B12 deficiency among vegetarians? Nutr Rev 71, 110117.CrossRefGoogle ScholarPubMed
Bakaloudi, DR, Halloran, A, Rippin, HL et al. (2021) Intake and adequacy of the vegan diet. A systematic review of the evidence. Clin Nutr 40, 35033521.CrossRefGoogle ScholarPubMed
Kristensen, NB, Madsen, ML, Hansen, TH et al. (2015) Intake of macro-and micronutrients in Danish vegans. Nutr J 14, 110.CrossRefGoogle ScholarPubMed
Weder, S, Keller, M, Fischer, M et al. (2022) Intake of micronutrients and fatty acids of vegetarian, vegan, and omnivorous children (1–3 years) in Germany (VeChi Diet Study). Eur J Nutr 61, 15071520.CrossRefGoogle ScholarPubMed
Benham, AJ, Gallegos, D, Hanna, KL, Hannan-Jones, MT (2021) Intake of vitamin B12 and other characteristics of women of reproductive age on a vegan diet in Australia. Public Health Nutr 24, 43974407.CrossRefGoogle ScholarPubMed
Schüpbach, R, Wegmüller, R, Berguerand, C et al. (2017) Micronutrient status and intake in omnivores, vegetarians and vegans in Switzerland. Eur J Nutr 56, 283293.CrossRefGoogle ScholarPubMed
Selinger, E, Kühn, T, Procházková, M et al. (2019) Vitamin B12 deficiency is prevalent among Czech vegans who do not use vitamin B12 supplements. Nutrients 11, 3019.CrossRefGoogle Scholar
Woo, KS, Kwok, TC, Celermajer, DS (2014) Vegan diet, subnormal vitamin B-12 status and cardiovascular health. Nutrients 6, 32593273.CrossRefGoogle ScholarPubMed
Lederer, A-K, Hannibal, L, Hettich, M et al. (2019) Vitamin B12 status upon short-term intervention with a vegan diet—a randomized controlled trial in healthy participants. Nutrients 11, 2815.CrossRefGoogle ScholarPubMed
White, ND (2022) Vitamin B12 and plant-predominant diets. Am J Lifestyle Med 16, 295297.CrossRefGoogle ScholarPubMed
Pawlak, R, Lester, S, Babatunde, T (2014) The prevalence of cobalamin deficiency among vegetarians assessed by serum vitamin B12: a review of literature. Eur J Clin Nutr 68, 541548.CrossRefGoogle ScholarPubMed
Fayet, F, Flood, V, Petocz, P, Samman, S (2014) Avoidance of meat and poultry decreases intakes of omega-3 fatty acids, vitamin B 12, selenium and zinc in young women. J Hum Nutr Diet 27, 135142.CrossRefGoogle ScholarPubMed
Seves, SM, Verkaik-Kloosterman, J, Biesbroek, S, Temme, EH (2017) Are more environmentally sustainable diets with less meat and dairy nutritionally adequate? Public Health Nutr 20, 20502062.CrossRefGoogle ScholarPubMed
Vatanparast, H, Islam, N, Shafiee, M, Ramdath, DD (2020) Increasing plant-based meat alternatives and decreasing red and processed meat in the diet differentially affect the diet quality and nutrient intakes of Canadians. Nutrients 12, 2034.CrossRefGoogle ScholarPubMed
Sivaprasad, M, Shalini, T, Balakrishna, N et al. (2016) Status of vitamin B12 and folate among the urban adult population in South India. Ann Nutr Metab 68, 94102.CrossRefGoogle ScholarPubMed
Dawczynski, C, Weidauer, T, Richert, C et al. (2022 ) Nutrient intake and nutrition status in vegetarians and vegans in comparison to omnivores – the Nutritional Evaluation (NuEva) study. Front Nutr 9, 819106.CrossRefGoogle ScholarPubMed
Agnoli, C, Baroni, L, Bertini, I et al. (2017) Position paper on vegetarian diets from the working group of the Italian Society of Human Nutrition. Nutr Metab Cardiovasc Dis 27, 10371052.CrossRefGoogle Scholar
Neufingerl, N, Eilander, A (2022) Nutrient intake and status in adults consuming plant-based diets compared to meat-eaters: a systematic review. Nutrients 14, 29.CrossRefGoogle Scholar
Niklewicz, A, Smith, AD, Smith, A et al. (2023) The importance of vitamin B12 for individuals choosing plant-based diets. Eur J Nutr 62, 15511559.CrossRefGoogle ScholarPubMed
Pulz, O, Gross, W (2004) Valuable products from biotechnology of microalgae. Appl Microbiol Biotechnol 65, 635648.CrossRefGoogle ScholarPubMed
Lafarga, T, Fernández-Sevilla, JM, González-López, C, Acién-Fernández, FG (2020) Spirulina for the food and functional food industries. Food Res Int 137, 109356.CrossRefGoogle ScholarPubMed
Watanabe, F, Katsura, H, Takenaka, S et al. (1999) Pseudovitamin B12 is the predominant cobamide of an algal health food, spirulina tablets. J Agric Food Chem 47, 47364741.CrossRefGoogle ScholarPubMed
Bito, T, Okumura, E, Fujishima, M, Watanabe, F (2020) Potential of Chlorella as a dietary supplement to promote human health. Nutrients 12, 2524.CrossRefGoogle ScholarPubMed
Sandgruber, F, Gielsdorf, A, Baur, AC et al. (2021) Variability in macro-and micronutrients of 15 commercially available microalgae powders. Mar Drugs 19, 310.CrossRefGoogle ScholarPubMed
Baroni, L, Scoglio, S, Benedetti, S et al. (2009) Effect of a Klamath algae product (“AFA-B12”) on blood levels of vitamin B12 and homocysteine in vegan subjects: a pilot study. Int J Vitam Nutr Res 79, 117.CrossRefGoogle ScholarPubMed
Miyamoto, E, Tanioka, Y, Nakao, T et al. (2006) Purification and characterization of a corrinoid-compound in an edible cyanobacterium Aphanizomenon flos-aquae as a nutritional supplementary food. J Agric Food Chem 54, 96049607.CrossRefGoogle Scholar
Behrendt, I (2017) Chlorella vulgaris – eine pflanzliche Vitamin B12 - Quelle für Vegetarier und Veganer? In 54 Wissenschaftlichen Kongress, vol. 23 [OS Wittek, I.; Norman, B. and Hahn, A., editor]: Deutsche Gesellschaft für Ernährung e.V.. https://www.dge.de/fileadmin/public/doc/wk/2017/DGE-Proc-Germ-Nutr-Soc-Vol-23-2017.pdf.Google Scholar
Castillejo, N, Martínez-Hernández, GB, Goffi, V et al. (2018) Natural vitamin B12 and fucose supplementation of green smoothies with edible algae and related quality changes during their shelf life. J Sci Food Agric 98, 24112421.CrossRefGoogle ScholarPubMed
Cornish, ML, Critchley, AT, Mouritsen, OG (2015) A role for dietary macroalgae in the amelioration of certain risk factors associated with cardiovascular disease. Phycologia 54, 649666.CrossRefGoogle Scholar
Kwak, C-S, Hwang, J-Y, Watanabe, F, Park, S-C (2008) Vitamin B12 contents in some Korean fermented foods and edible seaweeds. J Nutr Health 41, 439447.Google Scholar
Chandra-Hioe, MV, Lee, C, Arcot, J (2019) What is the cobalamin status among vegetarians and vegans in Australia? Int J Food Sci Nutr 70, 875886.CrossRefGoogle ScholarPubMed
Cao, J, Wang, J, Wang, S, Xu, X (2016) Porphyra species: a mini-review of its pharmacological and nutritional properties. J Med Food 19, 111119.CrossRefGoogle ScholarPubMed
Demarco, M, de Moraes, JO, Matos, ÂP et al. (2022) Digestibility, bioaccessibility and bioactivity of compounds from algae. Trends Food Sci Technol 121, 114128.CrossRefGoogle Scholar
Marczykowski, F, Breidenassel, C (2017) Vegan diet: reaching the reference values for nutrient intake of critical nutrients. Assortment and necessity of fortified foods. Ernahrungs Umschau 64, 210.Google Scholar
Green, R, Allen, LH, Bjørke-Monsen, A-L et al. (2017) Vitamin B12 deficiency. Nat Rev Dis Primers 3, 17040.CrossRefGoogle Scholar
Jedut, P, Szwajgier, D, Glibowski, P, Iłowiecka, K (2021) Some plant food products present on the polish market are a source of Vitamin B12. Appl Sci 11, 3601.CrossRefGoogle Scholar
Sánchez-Parra, E, Boutarfa, S, Aboal, M (2020) Are cyanotoxins the only toxic compound potentially present in microalgae supplements? Results from a study of ecological and non-ecological products. Toxins 12, 552.CrossRefGoogle ScholarPubMed
Rzymski, P, Niedzielski, P, Kaczmarek, N et al. (2015) The multidisciplinary approach to safety and toxicity assessment of microalgae-based food supplements following clinical cases of poisoning. Harmful Algae 46, 3442.CrossRefGoogle Scholar
Rzymski, P, Jaśkiewicz, M (2017) Microalgal food supplements from the perspective of Polish consumers: patterns of use, adverse events, and beneficial effects. J Appl Phycol 29, 18411850.CrossRefGoogle ScholarPubMed
Grosshagauer, S, Kraemer, K, Somoza, V (2020) The true value of spirulina. J Agric Food Chem 68, 41094115.CrossRefGoogle ScholarPubMed
Ampofo, J, Abbey, L (2022) Microalgae: bioactive composition, health benefits, safety and prospects as potential high-value ingredients for the functional food industry. Foods 11, 1744.CrossRefGoogle ScholarPubMed
Van Hassel, WHR, Ahn, A-C, Huybrechts, B et al. (2022) LC-MS/MS validation and quantification of cyanotoxins in algal food supplements from the Belgium market and their molecular origins. Toxins 14, 513.CrossRefGoogle ScholarPubMed
Muñoz, L, Díaz, I, Nelson, F (2020) Minerals in edible seaweed: health benefits and food safety issues. Crit Rev Food Sci Nutr 62, 15921607.CrossRefGoogle Scholar
Banach, JL, Koch, SJ, Hoffmans, Y, van den Burg, SW (2022) Seaweed value chain stakeholder perspectives for food and environmental safety hazards. Foods 11, 1514.CrossRefGoogle ScholarPubMed
Mendes, MC, Navalho, S, Ferreira, A et al. (2022) Algae as food in Europe: an overview of species diversity and their application. Foods 11, 1871.CrossRefGoogle ScholarPubMed
Rose, M, Lewis, J, Langford, N et al. (2007) Arsenic in seaweed—forms, concentration and dietary exposure. Food Chem Toxicol 45, 12631267.CrossRefGoogle ScholarPubMed
Raab, A, Stiboller, M, Gajdosechova, Z et al. (2016) Element content and daily intake from dietary supplements (nutraceuticals) based on algae, garlic, yeast fish and krill oils—should consumers be worried? J Food Compost Anal 53, 4960.CrossRefGoogle Scholar
Cho, TJ, Rhee, MS (2019) Health functionality and quality control of laver (Porphyra, Pyropia): current issues and future perspectives as an edible seaweed. Mar Drugs 18, 14.CrossRefGoogle ScholarPubMed
Vellinga, RE, Sam, M, Verhagen, H et al. (2022) Increasing seaweed consumption in the Netherlands and Portugal and the consequences for the intake of iodine, sodium, and exposure to chemical contaminants: a risk-benefit study. Front Nutr 8, 1042.CrossRefGoogle ScholarPubMed
Lomartire, S, Gonçalves, AM (2022) An overview of potential seaweed-derived bioactive compounds for pharmaceutical applications. Mar Drugs 20, 141.CrossRefGoogle ScholarPubMed
Circuncisão, AR, Catarino, MD, Cardoso, SM, Silva, AM (2018) Minerals from macroalgae origin: health benefits and risks for consumers. Mar Drugs 16, 400.CrossRefGoogle ScholarPubMed
Aakre, I, Solli, DD, Markhus, MW et al. (2021) Commercially available kelp and seaweed products – valuable iodine source or risk of excess intake? Food Nutr Res 65.CrossRefGoogle ScholarPubMed
Teas, J, Pino, S, Critchley, A, Braverman, LE (2004) Variability of iodine content in common commercially available edible seaweeds. Thyroid 14, 836841.CrossRefGoogle ScholarPubMed
Al-Dhabi, NA (2013) Heavy metal analysis in commercial spirulina products for human consumption. Saudi J Biol Sci 20, 383388.CrossRefGoogle ScholarPubMed
Muys, M, Sui, Y, Schwaiger, B et al. (2019) High variability in nutritional value and safety of commercially available Chlorella and Spirulina biomass indicates the need for smart production strategies. Bioresour Technol 275, 247257.CrossRefGoogle ScholarPubMed
European Commission (2018) Commission recommendation (EU) 2018/464 of 19 March 2018 on the monitoring of metals and iodine in seaweed, halophytes and products based on seaweed. Official J Eur Union 78, 1618.Google Scholar
Bundesinstitut für Risikobewertung (BfR) (2007) Gesundheitliche Risiken durch zu hohen Jodgehalt in getrockneten Algen. Aktualisierte Stellungnahme.Google Scholar
National Food Institute TUoD, Denmark, Sá Monteiro, M, Sloth, J et al. (2019) Analysis and risk assessment of seaweed. EFSA J 17, e170915.Google ScholarPubMed
Lähteenmäki-Uutela, A, Rahikainen, M, Camarena-Gómez, MT et al. (2021) European Union legislation on macroalgae products. Aquac Nutr 29, 487509.Google Scholar
Marles, RJ, Barrett, ML, Barnes, J et al. (2011) United States pharmacopeia safety evaluation of Spirulina. Crit Rev Food Sci Nutr 51, 593604.CrossRefGoogle ScholarPubMed
Li, M-Y, Wang, P, Wang, J-Y et al. (2019) Arsenic concentrations, speciation, and localization in 141 cultivated market mushrooms: implications for arsenic exposure to humans. Environ Sci Technol 53, 503511.CrossRefGoogle ScholarPubMed
Nearing, MM, Koch, I, Reimer, KJ (2014) Arsenic speciation in edible mushrooms. Environ Sci Technol 48, 1420314210.CrossRefGoogle ScholarPubMed
Llorente-Mirandes, T, Barbero, M, Rubio, R, López-Sánchez, JF (2014) Occurrence of inorganic arsenic in edible shiitake (Lentinula edodes) products. Food Chem 158, 207215.CrossRefGoogle ScholarPubMed
Zou, H, Zhou, C, Li, Y et al. (2019) Occurrence, toxicity, and speciation analysis of arsenic in edible mushrooms. Food Chem 281, 269284.CrossRefGoogle ScholarPubMed
Seyfferth, AL, McClatchy, C, Paukett, M (2016) Arsenic, lead, and cadmium in US mushrooms and substrate in relation to dietary exposure. Environ Sci Technol 50, 96619670.CrossRefGoogle ScholarPubMed
Nowakowski, P, Markiewicz-Żukowska, R, Soroczyńska, J et al. (2021) Evaluation of toxic element content and health risk assessment of edible wild mushrooms. J Food Compost Anal 96, 103698.CrossRefGoogle Scholar
Ab Rhaman, SMS, Naher, L, Siddiquee, S (2021) Mushroom quality related with various substrates’ bioaccumulation and translocation of heavy metals. J Fungi (Basel) 8, 42.CrossRefGoogle ScholarPubMed
Falandysz, J, Borovička, J (2013) Macro and trace mineral constituents and radionuclides in mushrooms: health benefits and risks. Appl Microbiol Biotechnol 97, 477501.CrossRefGoogle ScholarPubMed
Dowlati, M, Sobhi, HR, Esrafili, A et al. (2021) Heavy metals content in edible mushrooms: a systematic review, meta-analysis and health risk assessment. Trends Food Sci Technol 109, 527535.CrossRefGoogle Scholar
El-Ramady, H, Llanaj, X, Prokisch, J (2021) Edible mushroom cultivated in polluted soils and its potential risks on human health: a short communication. Egypt J Soil Sci 61, 110.CrossRefGoogle Scholar
Duff, MC, Ramsey, ML (2008) Accumulation of radiocesium by mushrooms in the environment: a literature review. J Environ Radioact 99, 912932.CrossRefGoogle ScholarPubMed
Ronda, O, Grządka, E, Ostolska, I et al. (2022) Accumulation of radioisotopes and heavy metals in selected species of mushrooms. Food Chem 367, 130670.CrossRefGoogle ScholarPubMed
Strumińska-Parulska, D, Falandysz, J, Moniakowska, A (2021) Beta-emitting radionuclides in wild mushrooms and potential radiotoxicity for their consumers. Trends Food Sci Technol 114, 672683.CrossRefGoogle Scholar
Guillén, J, Baeza, A (2014) Radioactivity in mushrooms: a health hazard? Food Chem 154, 1425.CrossRefGoogle ScholarPubMed
EFSA (2021) Safety of Wolffia globosa powder as a novel food pursuant to Regulation (EU) 2015/2283. EFSA J 19, e06938.Google Scholar
Liem, I, Steinkraus, K, Cronk, T (1977) Production of vitamin B-12 in tempeh, a fermented soybean food. Appl Environ Microbiol 34, 773776.CrossRefGoogle ScholarPubMed
Mo, H, Kariluoto, S, Piironen, V et al. (2013) Effect of soybean processing on content and bioaccessibility of folate, vitamin B12 and isoflavones in tofu and tempe. Food Chem 141, 24182425.CrossRefGoogle ScholarPubMed
Masuda, M, Ide, M, Utsumi, H et al. (2012) Production potency of folate, vitamin B12, and thiamine by lactic acid bacteria isolated from Japanese pickles. Biosci Biotechnol Biochem 76, 20612067.CrossRefGoogle ScholarPubMed
Zugravu, C-A, Macri, A, Belc, N, Bohiltea, R (2021) Efficacy of supplementation with methylcobalamin and cyancobalamin in maintaining the level of serum holotranscobalamin in a group of plant-based diet (vegan) adults. Exp Ther Med 22, 17.CrossRefGoogle Scholar
Brown, DD (2018) Nutritional considerations for the vegetarian and vegan dancer. J Dance Med Sci 22, 4453.CrossRefGoogle ScholarPubMed
Richter, M, Boeing, H, Grünewald-Funk, D et al. (2016) Vegan diet. Position of the German Nutrition Society (DGE). Ernahrungs Umschau 63, 92102.Google Scholar
Schwarz, J, Dschietzig, T, Schwarz, J et al. (2014) The influence of a whole food vegan diet with Nori algae and wild mushrooms on selected blood parameters. Clin Lab 60, 20392050.CrossRefGoogle ScholarPubMed
Strain, J, Hughes, C, Pentieva, K et al. (2017) The B-Vitamins. In Sustainable Nutrition in a Changing World, 1st ed., pp. 185205 [Biesalski, HK, Drewnowski, A, Dwyer, JT, Strain, J, Weber, P and Eggersdorfer, M, editors] Cham, Switzerland: Springer International Publishing AG.CrossRefGoogle Scholar
Yamada, Yamada, Fukuda, Yamada (1999) Bioavailability of dried asakusanori (Porphyra tenera) as a source of cobalamin (vitamin B12). Int J Vitam Nutr Res 69, 412418.CrossRefGoogle Scholar
Gehring, J, Touvier, M, Baudry, J et al. (2021) Consumption of ultra-processed foods by pesco-vegetarians, vegetarians, and vegans: associations with duration and age at diet initiation. J Nutr 151, 120131.CrossRefGoogle ScholarPubMed
Gibney, MJ (2021) Food technology and plant-based diets. J Nutr 151, 12.CrossRefGoogle Scholar
Satija, A, Bhupathiraju, SN, Spiegelman, D et al. (2017) Healthful and unhealthful plant-based diets and the risk of coronary heart disease in US adults. J Am Coll Cardiol 70, 411422.CrossRefGoogle Scholar
Wang, F, Ugai, T, Haruki, K et al. (2022) Healthy and unhealthy plant-based diets in relation to the incidence of colorectal cancer overall and by molecular subtypes. Clin Transl Med 12, e893.CrossRefGoogle ScholarPubMed
Williams, KA, Patel, H (2017) Healthy plant-based diet: what does it really mean? Washington, DC: American College of Cardiology Foundation, vol. 70. 423425.Google ScholarPubMed
Pointke, M, Pawelzik, E (2022) Plant-based alternative products: are they healthy alternatives? Micro- and macronutrients and nutritional scoring. Nutrients 14, 601.CrossRefGoogle ScholarPubMed
Hemler, EC, Hu, FB (2019) Plant-based diets for cardiovascular disease prevention: all plant foods are not created equal. Curr Atheroscler Rep 21, 18.CrossRefGoogle Scholar
Khandpur, N, Martinez-Steele, E, Sun, Q (2021) Plant-based meat and dairy substitutes as appropriate alternatives to animal-based products? J Nutr 151, 34.CrossRefGoogle ScholarPubMed
D’Alessandro, C, Pezzica, J, Bolli, C et al. (2022) Processed plant-based foods for CKD patients: good choice, but be aware. Int J Environ Res Public Health 19, 6653.CrossRefGoogle ScholarPubMed
Orlich, MJ, Sabaté, J, Mashchak, A et al. (2022) Ultra-processed food intake and animal-based food intake and mortality in the Adventist Health Study-2. Am J Clin Nutr 115, 15891601.CrossRefGoogle Scholar
Craig, WJ, Mangels, AR, Fresán, U et al. (2021) The safe and effective use of plant-based diets with guidelines for health professionals. Nutrients 13, 4144.CrossRefGoogle ScholarPubMed
Craig, WJ, Mangels, AR (2009) Position of the American Dietetic Association: vegetarian diets. J Am Diet Assoc 109, 1266.Google ScholarPubMed
Alcorta, A, Porta, A, Tárrega, A et al. (2021) Foods for plant-based diets: challenges and innovations. Foods 10, 293.CrossRefGoogle ScholarPubMed
Ströhle, A, Löser, C, Behrendt, I et al. (2018) Alternative ernährungsformen: allgemeine aspekte und vegetarische kostformen. Rehabilitation 57, 5570.Google Scholar
Hever, J (2016) Plant-based diets: a physician’s guide. Perm J 20.CrossRefGoogle ScholarPubMed
Jakše, B (2021) Placing a well-designed vegan diet for slovenes. Nutrients 13, 4545.CrossRefGoogle ScholarPubMed
Mangels, AR, Messina, V, Melina, V (2003) Position of the American Dietetic Association and Dietitians of Canada: vegetarian diets. J Am Diet Assoc 103, 748766.Google Scholar
Liebman, SE, Joshi, S (2022) Plant-based diets and peritoneal dialysis: a review. Nutrients 14, 1304.CrossRefGoogle ScholarPubMed
Parra-Soto, S, Ahumada, D, Petermann-Rocha, F et al. (2022) Association of meat, vegetarian, pescatarian and fish-poultry diets with risk of 19 cancer sites and all cancer: findings from the UK Biobank prospective cohort study and meta-analysis. BMC Med 20, 116.CrossRefGoogle Scholar
Boushey, C, Ard, J, Bazzano, L et al. (2020) What is the relationship between dietary patterns consumed and all-cause mortality? In Dietary Patterns and All-Cause Mortality: A Systematic Review [Internet]: USDA Nutrition Evidence Systematic Review.CrossRefGoogle Scholar
Selinger, E, Neuenschwander, M, Koller, A et al. (2022) Evidence of a vegan diet for health benefits and risks–an umbrella review of meta-analyses of observational and clinical studies. Crit Rev Food Sci Nutr 63, 111.Google Scholar
Tahreem, A, Rakha, A, Rabail, R et al. (2022) Fad diets: facts and fiction. Front Nutr 9, 960922.CrossRefGoogle ScholarPubMed
Appleby, PN, Crowe, FL, Bradbury, KE et al. (2016) Mortality in vegetarians and comparable nonvegetarians in the United Kingdom. Am J Clin Nutr 103, 218230.CrossRefGoogle ScholarPubMed
Rocha, JP, Laster, J, Parag, B, Shah, NU (2019) Multiple health benefits and minimal risks associated with vegetarian diets. Curr Nutr Rep 8, 374381.CrossRefGoogle ScholarPubMed
Kim, H, Caulfield, LE, Garcia-Larsen, V et al. (2019) Plant-based diets are associated with a lower risk of incident cardiovascular disease, cardiovascular disease mortality, and all-cause mortality in a general population of middle-aged adults. J Am Heart Assoc 8, e012865.CrossRefGoogle Scholar
Szabo, Z, Koczka, V, Marosvolgyi, T et al. (2021) Possible biochemical processes underlying the positive health effects of plant-based diets—a narrative review. Nutrients 13, 2593.CrossRefGoogle ScholarPubMed
Zhao, Y, Zhan, J, Wang, Y, Wang, D (2022) The relationship between plant-based diet and risk of digestive system cancers: a meta-analysis based on 3,059,009 subjects. Front Public Health 10, 1596.Google ScholarPubMed
Marrone, G, Guerriero, C, Palazzetti, D et al. (2021) Vegan diet health benefits in metabolic syndrome. Nutrients 13, 817.CrossRefGoogle ScholarPubMed
Dybvik, JS, Svendsen, M, Aune, D (2022) Vegetarian and vegan diets and the risk of cardiovascular disease, ischemic heart disease and stroke: a systematic review and meta-analysis of prospective cohort studies. Eur J Nutr 62, 119.Google ScholarPubMed
Kahleova, H, Levin, S, Barnard, ND (2018) Vegetarian dietary patterns and cardiovascular disease. Prog Cardiovasc Dis 61, 5461.CrossRefGoogle ScholarPubMed
Chauveau, P, Koppe, L, Combe, C et al. (2019) Vegetarian diets and chronic kidney disease. Nephrol Dial Transplant 34, 199207.CrossRefGoogle ScholarPubMed
Huang, R-Y, Huang, C-C, Hu, FB, Chavarro, JE (2016) Vegetarian diets and weight reduction: a meta-analysis of randomized controlled trials. J Gen Intern Med 31, 109116.CrossRefGoogle ScholarPubMed
Leitzmann, C (2014) Vegetarian nutrition: past, present, future. Am J Clin Nutr 100, 496S502S.CrossRefGoogle ScholarPubMed
Dinu, M, Abbate, R, Gensini, GF et al. (2017) Vegetarian, vegan diets and multiple health outcomes: a systematic review with meta-analysis of observational studies. Crit Rev Food Sci Nutr 57, 36403649.CrossRefGoogle ScholarPubMed
Nhan, J, Sgambat, K, Moudgil, A (2023) Plant-based diets: a fad or the future of medical nutrition therapy for children with chronic kidney disease? Pediatr Nephrol 38, 113.CrossRefGoogle ScholarPubMed
Nebl, J, Schuchardt, JP, Ströhle, A et al. (2019) Micronutrient status of recreational runners with vegetarian or non-vegetarian dietary patterns. Nutrients 11, 1146.CrossRefGoogle ScholarPubMed
Weikert, C, Trefflich, I, Menzel, J et al. (2020) Vitamin and mineral status in a vegan diet. Dtsch Arztebl Int 117, 575.Google ScholarPubMed
Donaldson, MS (2000) Metabolic vitamin B12 status on a mostly raw vegan diet with follow-up using tablets, nutritional yeast, or probiotic supplements. Ann Nutr Metab 44, 229234.CrossRefGoogle ScholarPubMed
Damayanti, D, Jaceldo-Siegl, K, Beeson, WL et al. (2018) Foods and supplements associated with vitamin B12 biomarkers among vegetarian and non-vegetarian participants of the Adventist Health Study-2 (AHS-2) calibration study. Nutrients 10, 722.CrossRefGoogle ScholarPubMed
Gallego-Narbón, A, Zapatera, B, Álvarez, I, Vaquero, MP (2018) Methylmalonic acid levels and their relation with cobalamin supplementation in Spanish vegetarians. Plant Foods Hum Nutr 73, 166171.CrossRefGoogle ScholarPubMed
Gallego-Narbón, A, Zapatera, B, Barrios, L, Vaquero, MP (2019) Vitamin B12 and folate status in Spanish lacto-ovo vegetarians and vegans. J Nutr Sci 8, e7.CrossRefGoogle ScholarPubMed
Del Bo, C, Riso, P, Gardana, C et al. (2019) Effect of two different sublingual dosages of vitamin B12 on cobalamin nutritional status in vegans and vegetarians with a marginal deficiency: a randomized controlled trial. Clin Nutr 38, 575583.CrossRefGoogle ScholarPubMed
Alexy, U, Fischer, M, Weder, S et al. (2021) Nutrient intake and status of german children and adolescents consuming vegetarian, vegan or omnivore diets: results of the vechi youth study. Nutrients 13, 1707.CrossRefGoogle ScholarPubMed
Sutter, DO, Bender, N (2021) Nutrient status and growth in vegan children. Nutr Res 91, 1325.CrossRefGoogle ScholarPubMed
Venti, CA, Johnston, CS (2002) Modified food guide pyramid for lactovegetarians and vegans. J Nutr 132, 10501054.CrossRefGoogle ScholarPubMed
Weder, S, Schaefer, C, Keller, M (2018) The Gießen vegan food pyramid. Ernahrungs Umschau 65, 134143.Google Scholar
Karlsen, MC, Rogers, G, Miki, A et al. (2019) Theoretical food and nutrient composition of whole-food plant-based and vegan diets compared to current dietary recommendations. Nutrients 11, 625.CrossRefGoogle ScholarPubMed
Vollmer, I, Keller, M, Kroke, A (2018) Vegan diet: utilization of dietary supplements and fortified foods. An internet-based survey. Ernahrungs Umschau 65, 144153.Google Scholar
Jakše, B, Jakše, B, Godnov, U, Pinter, S (2021) Nutritional, cardiovascular health and lifestyle status of ‘health conscious’ adult vegans and non-vegans from Slovenia: a cross-sectional self-reported survey. Int J Environ Res Public Health 18, 5968.CrossRefGoogle ScholarPubMed
Jakše, B, Jakše, B, Pinter, S et al. (2021) Nutrient and food intake of participants in a whole-food plant-based lifestyle program. J Am Coll Nutr 40, 333348.CrossRefGoogle Scholar
Rudloff, S, Bührer, C, Jochum, F et al. (2019) Vegetarian diets in childhood and adolescence. Mol Cell Pediatr 6, 17.CrossRefGoogle ScholarPubMed
Richter, M, Kroke, A, Grünewald-Funk, D et al. (2020) Update to the position of the German Nutrition Society on vegan diets in population groups with special nutritional requirements. Position of the German Nutrition Society 2, 6472.Google Scholar
Baroni, L, Goggi, S, Battaglino, R et al. (2018) Vegan nutrition for mothers and children: practical tools for healthcare providers. Nutrients 11, 5.CrossRefGoogle ScholarPubMed
Baroni, L, Goggi, S, Battino, M (2018) VegPlate: a Mediterranean-based food guide for Italian adult, pregnant, and lactating vegetarians. J Acad Nutr Diet 118, 22352243.CrossRefGoogle ScholarPubMed
Baroni, L, Goggi, S, Battino, M (2019) Planning well-balanced vegetarian diets in infants, children, and adolescents: the vegplate junior. J Acad Nutr Diet 119, 10671074.CrossRefGoogle ScholarPubMed
Tso, R, Forde, CG (2021) Unintended consequences: nutritional impact and potential pitfalls of switching from animal-to plant-based foods. Nutrients 13, 2527.CrossRefGoogle ScholarPubMed
Mądry, E, Lisowska, A, Grebowiec, P (2012) The impact of vegan diet on B-12 status in healthy omnivores: five-year prospective study. Acta Sci Pol Technol Aliment 11, 209212.Google ScholarPubMed
Kiely, ME (2021) Risks and benefits of vegan and vegetarian diets in children. Proc Nutr Soc 80, 159164.CrossRefGoogle ScholarPubMed
Redecillas-Ferreiro, S, Moráis-López, A, Moreno-Villares, JM (2020) Position paper on vegetarian diets in infants and children. Committee on Nutrition and Breastfeeding of the Spanish Paediatric Association. An Pediatr (Engl Ed) 92, 306. e301e306.Google Scholar
Rashid, S, Meier, V, Patrick, H (2021) Review of vitamin B12 deficiency in pregnancy: a diagnosis not to miss as veganism and vegetarianism become more prevalent. Eur J Haematol 106, 450455.CrossRefGoogle Scholar
Reid, MA, Marsh, KA, Zeuschner, CL et al. (2013) Meeting the nutrient reference values on a vegetarian diet. Med J Aust 199, S33S40.CrossRefGoogle ScholarPubMed
Menal-Puey, S, Marques-Lopes, I (2016) Development of a food guide for the vegetarians of Spain. J Acad Nutr Diet 117, 15091516.CrossRefGoogle ScholarPubMed
Vincenti, A, Bertuzzo, L, Limitone, A et al. (2021) Perspective: practical approach to preventing subclinical b12 deficiency in elderly population. Nutrients 13, 1913.CrossRefGoogle ScholarPubMed
Storz, MA, Müller, A, Niederreiter, L et al. (2023) A cross-sectional study of nutritional status in healthy, young, physically-active German omnivores, vegetarians and vegans reveals adequate vitamin B12 status in supplemented vegans. Ann Med 55, 2269969.CrossRefGoogle ScholarPubMed
Hunt, A, Harrington, D, Robinson, S (2014) Vitamin B12 deficiency. BMJ 349.CrossRefGoogle ScholarPubMed
Berry Ottaway, P (2010) Stability of vitamins during food processing and storage. In Chemical Deterioration and Physical Instability of Food and Beverages, pp. 539560 [Skibsted, LH, Risbo, J and Andersen, ML, editors] Cambridge, UK: Woodhead Publishing.CrossRefGoogle Scholar
Godoy, HT, Amaya-Farfan, J, Rodriguez-Amaya, DB (2021) Degradation of vitamins. In Chemical Changes During Processing and Storage of Foods, pp. 329383 [Rodriguez-Amaya, DB and Amaya-Farfan, J, editors] Academic Press.CrossRefGoogle Scholar
Bergström, L (1994) Nutrient losses and gains in the preparation of foods. Rapport-Livsmedelsverket (Sweden).Google Scholar
Bennink, M, Ono, K (1982) Vitamin B12, E and D content of raw and cooked beef. J Food Sci 47, 17861792.CrossRefGoogle Scholar
Lee, W, Lee, Y-B, Huh, MH, Choi, JK (2022) Determination of the chemical stability of cyanocobalamin in medical food by a validated immunoaffinity column-linked HPLC method. J Food Qual 2022, 18.CrossRefGoogle Scholar
Ahmad, I, Qadeer, K, Zahid, S et al. (2014) Effect of ascorbic acid on the degradation of cyanocobalamin and hydroxocobalamin in aqueous solution: a kinetic study. AAPS PharmSciTech 15, 13241333.CrossRefGoogle ScholarPubMed
Henry, C, Heppell, N (2002) Nutritional losses and gains during processing: future problems and issues. Proc Nutr Soc 61, 145148.CrossRefGoogle ScholarPubMed
Yessaad, M, Bernard, L, Bourdeaux, D et al. (2018) Development of a stability indicating method for simultaneous analysis of five water-soluble vitamins by liquid chromatography. Pharm Technol Hosp Pharm 3, 207218.CrossRefGoogle Scholar
Johns, PW, Das, A, Kuil, EM et al. (2015) Cocoa polyphenols accelerate vitamin B 12 degradation in heated chocolate milk. Int J Food Sci Technol 50, 421430.CrossRefGoogle Scholar
Lie, AH, Chandra-Hioe, MV, Arcot, J (2019) Sorbitol enhances the physicochemical stability of B12 vitamers. Int J Vitam Nutr Res.Google Scholar
Okamoto, N, Bito, T, Hiura, N et al. (2020) Food additives (hypochlorous acid water, sodium metabisulfite, and sodium sulfite) strongly affect the chemical and biological properties of vitamin B12 in aqueous solution. ACS Omega 5, 62076214.CrossRefGoogle ScholarPubMed
Edelmann, M, Chamlagain, B, Santin, M et al. (2016) Stability of added and in situ-produced vitamin B12 in breadmaking. Food Chem 204, 2128.CrossRefGoogle ScholarPubMed
Fedosov, SN, Ruetz, M, Gruber, K et al. (2011) A blue corrinoid from partial degradation of vitamin B12 in aqueous bicarbonate: spectra, structure, and interaction with proteins of B12 transport. Biochemistry 50, 80908101.CrossRefGoogle ScholarPubMed
Bajaj, SR, Singhal, RS (2020) Degradation kinetics of vitamin B12 in model systems of different pH and extrapolation to carrot and lime juices. J Food Eng 272, 109800.CrossRefGoogle Scholar
Schnellbaecher, A, Binder, D, Bellmaine, S, Zimmer, A (2019) Vitamins in cell culture media: stability and stabilization strategies. Biotechnol Bioeng 116, 15371555.CrossRefGoogle ScholarPubMed
Yamada, K, Shimodaira, M, Chida, S et al. (2008) Degradation of vitamin B12 in dietary supplements. Int J Vitam Nutr Res 78, 195203.CrossRefGoogle ScholarPubMed
Modupe, O, Diosady, LL (2021) Quadruple fortification of salt for the delivery of iron, iodine, folic acid, and vitamin B12 to vulnerable populations. J Food Eng 300, 110525.CrossRefGoogle ScholarPubMed
Raju, CSK, Lee, LY, Schiel, JE, Long, SE (2013) A simple and sensitive LC-ICP-MS method for the accurate determination of vitamin B12 in fortified breakfast cereals and multivitamin tablets. J Anal At Spectrom 28, 901907.CrossRefGoogle Scholar
Temova Rakuša, Ž, Roškar, R, Hickey, N, Geremia, S (2022) Vitamin B12 in foods, food supplements, and medicines—a review of its role and properties with a focus on its stability. Molecules 28, 240.CrossRefGoogle ScholarPubMed
Campo, M, Muela, E, Olleta, J et al. (2013) Influence of cooking method on the nutrient composition of Spanish light lamb. J Food Compost Anal 31, 185190.CrossRefGoogle Scholar
Czerwonka, M, Szterk, A, Waszkiewicz-Robak, B (2014) Vitamin B12 content in raw and cooked beef. Meat Sci 96, 13711375.CrossRefGoogle ScholarPubMed
Rolls, BA, Porter, JWG (1973) Some effects of processing and storage on the nutritive value of milk and milk products. Proc Nutr Soc 32, 915.CrossRefGoogle ScholarPubMed
Scott, K, BISHOP, DR (1986) Nutrient content of milk and milk products: vitamins of the B complex and vitamin C in retail market milk and milk products. Int J Dairy Technol 39, 3235.CrossRefGoogle Scholar
Ortigues-Marty, I, Thomas, E, Prévéraud, D et al. (2006) Influence of maturation and cooking treatments on the nutritional value of bovine meats: water losses and vitamin B12. Meat Sci 73, 451458.CrossRefGoogle ScholarPubMed
Nishioka, M, Kanosue, F, Yabuta, Y, Watanabe, F (2011) Loss of vitamin B12 in fish (round herring) meats during various cooking treatments. J Nutr Sci Vitaminol 57, 432436.CrossRefGoogle ScholarPubMed
Riccio, F, Mennella, C, Fogliano, V (2006) Effect of cooking on the concentration of Vitamins B in fortified meat products. J Pharm Biomed Anal 41, 15921595.CrossRefGoogle ScholarPubMed
Van Heerden, S, Schönfeldt, H, Smith, M, van Rensburg, DJ (2002) Nutrient content of South African chickens. J Food Compost Anal 15, 4764.CrossRefGoogle Scholar
Severi, S, Bedogni, G, Manzieri, AM et al. (1997) Effects of cooking and storage methods on the micronutrient content of foods. Eur J Cancer Prev 6, S21S24.CrossRefGoogle ScholarPubMed
Bognár, A (2002) Tables on weight yield of food and retention factors of food constituents for the calculation of nutrient composition of cooked foods (dishes). Karlsruhe, Germany: Bundesforschungsanstalt für Ernährung.Google Scholar
Bell, S, Becker, W, Vásquez-Caicedo, A et al. (2006) Report on nutrient losses and gains factors used in European food composition databases. Karlsruhe, Germany: Federal Research Centre for Nutrition and Food.Google Scholar
Öhrvik, V, Carlsen, MH, Källman, A, Martinsen, TA (2015) Improving food composition data by standardizing calculation methods, TemaNord. Copenhagen: Nordisk Ministerråd.Google Scholar
Oamen, E, Hansen, A, Swartzel, K (1989) Effect of ultra-high temperature steam injection processing and aseptic storage on labile water-soluble vitamins in milk. J Dairy Sci 72, 614619.CrossRefGoogle Scholar
Amador-Espejo, G, Gallardo-Chacon, J, Nykänen, H et al. (2015) Effect of ultra high-pressure homogenization on hydro-and liposoluble milk vitamins. Food Res Int 77, 4954.CrossRefGoogle Scholar
Andersson, I, Öste, R (1994) Nutritional quality of pasteurized milk. Vitamin B12, folate and ascorbic acid content during storage. Int Dairy J 4, 161172.CrossRefGoogle Scholar
Arkbåge, K, Witthöft, C, Fondén, R, Jägerstad, M (2003) Retention of vitamin B12 during manufacture of six fermented dairy products using a validated radio protein-binding assay. Int Dairy J 13, 101109.CrossRefGoogle Scholar
Graham, DM (1974) Alteration of nutritive value resulting from processing and fortification of milk and milk products. J Dairy Sci 57, 738745.CrossRefGoogle ScholarPubMed
Lešková, E, Kubíková, J, Kováčiková, E et al. (2006) Vitamin losses: retention during heat treatment and continual changes expressed by mathematical models. J Food Compost Anal 19, 252276.CrossRefGoogle Scholar
Kyritsi, A, Tzia, C, Karathanos, VT (2011) Vitamin fortified rice grain using spraying and soaking methods. Lwt-Food Sci Technol 44, 312320.CrossRefGoogle Scholar
Steiger, G, Muller-Fischer, N, Cori, H, Conde-Petit, B (2014) Fortification of rice: technologies and nutrients. Ann N Y Acad Sci 1324, 2939.CrossRefGoogle ScholarPubMed
Sato, K, Kudo, Y, Muramatsu, K (2004) Incorporation of a high level of vitamin B12 into a vegetable, kaiware daikon (Japanese radish sprout), by the absorption from its seeds. Biochim Biophys Acta Gen Subj 1672, 135137.CrossRefGoogle ScholarPubMed
Watanabe, F, Abe, K, Fujita, T et al. (1998) Effects of microwave heating on the loss of vitamin B12 in foods. J Agric Food Chem 46, 206210.CrossRefGoogle ScholarPubMed
Tian, Y, Zhao, Y, Huang, J et al. (2016) Effects of different drying methods on the product quality and volatile compounds of whole shiitake mushrooms. Food Chem 197, 714722.CrossRefGoogle Scholar
Zheng, Y, Xiang, S, Zhang, H et al. (2021) Vitamin B12 enriched in spinach and its effects on gut microbiota. J Agric Food Chem 69, 22042212.CrossRefGoogle ScholarPubMed
Bajaj, SR, Singhal, RS (2019) Effect of extrusion processing and hydrocolloids on the stability of added vitamin B12 and physico-functional properties of the fortified puffed extrudates. Lwt-Food Sci Technol 101, 3239.CrossRefGoogle Scholar
Chamlagain, B (2016) Fermentation fortification of active vitamin B12 in food matrices using Propionibacterium freudenreichii: analysis, production and stability.CrossRefGoogle Scholar
Bajaj, SR, Singhal, RS (2021) Fortification of wheat flour and oil with vitamins B12 and D3: effect of processing and storage. J Food Compost Anal 96, 103703.CrossRefGoogle Scholar
Juzeniene, A, Nizauskaite, Z (2013) Photodegradation of cobalamins in aqueous solutions and in human blood. J Photochem Photobiol B 122, 714.CrossRefGoogle ScholarPubMed
Bito, T, Ohishi, N, Hatanaka, Y et al. (2013) Production and characterization of cyanocobalamin-enriched lettuce (Lactuca sativa L.) grown using hydroponics. J Agric Food Chem 61, 38523858.CrossRefGoogle ScholarPubMed
Watanabe, F, Katsura, H, Abe, K, Nakano, Y (2000) Effect of light-induced riboflavin degradation on the loss of cobalamin in milk. J Home Econ Jpn 51, 231234.Google Scholar
Saffert, A, Pieper, G, Jetten, J (2006) Effect of package light transmittance on the vitamin content of pasteurized whole milk. Packag Technol Sci 19, 211218.CrossRefGoogle Scholar
Campos-Gimnez, E, Fontannaz, P, Trisconi, M-J et al. (2008) Determination of vitamin B12 in food products by liquid chromatography/UV detection with immunoaffinity extraction: single-laboratory validation. J AOAC Int 91, 786793.CrossRefGoogle Scholar
Wang, H, Shou, Y, Zhu, X et al. (2019) Stability of vitamin B12 with the protection of whey proteins and their effects on the gut microbiome. Food Chem 276, 298306.CrossRefGoogle ScholarPubMed
Repossi, A, Zironi, E, Gazzotti, T et al. (2017) Vitamin B12 determination in milk, whey and different by-products of ricotta cheese production by ultra performance liquid chromatography coupled with tandem mass spectrometry. Ital J Food Saf 6, 152155.Google ScholarPubMed
Rabah, H, Carmo, FLRD, Jan, G (2017) Dairy propionibacteria: versatile probiotics. Microorganisms 5, 24.CrossRefGoogle ScholarPubMed
Poonam, Pophaly SD, Tomar, SK et al. (2012) Multifaceted attributes of dairy propionibacteria: a review. World J Microbiol Biotechnol 28, 30813095.CrossRefGoogle ScholarPubMed
Kilcast, D (1994) Effect of irradiation on vitamins. Food Chem 49, 157164.CrossRefGoogle Scholar
Woodside, J (2015) Nutritional aspects of irradiated food. Stewart Postharvest Rev 11, 16.CrossRefGoogle Scholar
Fox, JB Jr, Thayer, DW, Jenkins, RK et al. (1989) Effect of gamma irradiation on the B vitamins of pork chops and chicken breasts. Int J Radiat Biol 55, 689703.CrossRefGoogle Scholar
Takenaka, S, Sugiyama, S, Watanabe, F et al. (1997) Effects of carnosine and anserine on the destruction of vitamin B12 with vitamin C in the presence of copper. Biosci Biotechnol Biochem 61, 21372139.CrossRefGoogle ScholarPubMed
Ford, JE, Hurrell, RF, Finot, PA (1983) Storage of milk powders under adverse conditions. 2. Influence on the content of water-soluble vitamins. Br J Nutr 49, 355364.CrossRefGoogle ScholarPubMed
Hemery, YM, Fontan, L, Laillou, A et al. (2020) Influence of storage conditions and packaging of fortified wheat flour on microbial load and stability of folate and vitamin B12. Food Chem X 5, 100076.CrossRefGoogle ScholarPubMed
Zwart, S, Kloeris, V, Perchonok, M et al. (2009) Assessment of nutrient stability in foods from the space food system after long-duration spaceflight on the ISS. J Food Sci 74, H209H217.CrossRefGoogle ScholarPubMed
Bajaj, SR, Singhal, RS (2021) Enhancement of stability of vitamin B12 by co-crystallization: a convenient and palatable form of fortification. J Food Eng 291, 110231.CrossRefGoogle Scholar
Vinodkumar, M, Rajagopalan, S (2009) Multiple micronutrient fortification of salt. Eur J Clin Nutr 63, 437445.CrossRefGoogle Scholar
Obeid, R, Fedosov, SN, Nexo, E (2015) Cobalamin coenzyme forms are not likely to be superior to cyano- and hydroxyl-cobalamin in prevention or treatment of cobalamin deficiency. Mol Nutr Food Res 59, 13641372.CrossRefGoogle ScholarPubMed
Stabler, SP (2013) Vitamin B12 deficiency. N Engl J Med 368, 149160.CrossRefGoogle ScholarPubMed
EFSA (2018) Safety and efficacy of vitamin B12 (in the form of cyanocobalamin) produced by Ensifer spp. as a feed additive for all animal species based on a dossier submitted by VITAC EEIG. EFSA J 16, e05336.Google Scholar
EFSA (2020) Safety of vitamin B12 (in the form of cyanocobalamin) produced by Ensifer adhaerens CNCM-I 5541 for all animal species. EFSA J 18, e06335.Google Scholar
EU Parliament (2006) Regulation (EC) No 1925/2006 of the European Parliament and of the Council of 20 December 2006 on the addition of vitamins and minerals and of certain other substances to foods. Official J Eur Union 50, 2638.Google Scholar
Mahalle, N, Bhide, V, Greibe, E et al. (2019) Comparative bioavailability of synthetic B12 and dietary vitamin B12 present in cow and buffalo milk: a prospective study in lactovegetarian Indians. Nutrients 11, 304.CrossRefGoogle ScholarPubMed
EFSA (2006) Tolerable upper intake levels for vitamins and minerals.Google Scholar
Devi, S, Pasanna, RM, Shamshuddin, Z et al. (2020) Measuring vitamin B-12 bioavailability with [13C]-cyanocobalamin in humans. Am J Clin Nutr 112, 15041515.CrossRefGoogle ScholarPubMed
European Commission (2006) Commission Directive 2006/141/EC of 22 December 2006 on infant formulae and follow-on formulae and amending Directive 1999/21/EC. Official J Eur Union 49, 133.Google Scholar
European Commission (2006) Commission directive 2006/125/EC of 5 December 2006 on processed cereal-based foods and baby foods for infants and young children. Official J Eur Union L 339/16.Google Scholar
Calvillo, Á, Pellicer, T, Carnicer, M, Planas, A (2022) Bioprocess strategies for vitamin B12 production by microbial fermentation and its market applications. Bioengineering 9, 365.CrossRefGoogle ScholarPubMed
Thompson, JP, Marrs, TC (2012) Hydroxocobalamin in cyanide poisoning. Clin Toxicol 50, 875885.CrossRefGoogle ScholarPubMed
Fidaleo, M, Tacconi, S, Sbarigia, C et al. (2021) Current nanocarrier strategies improve vitamin B12 pharmacokinetics, ameliorate patients’ lives, and reduce costs. Nanomaterials 11, 743.CrossRefGoogle ScholarPubMed
Green, R (2017) Vitamin B12 deficiency from the perspective of a practicing hematologist. Blood 129, 26032611.CrossRefGoogle ScholarPubMed
Dharmarajan, TS, Norkus, EP (2001) Approaches to vitamin B12 deficiency: early treatment may prevent devastating complications. Postgrad Med 110, 99105.CrossRefGoogle Scholar
Carmel, R (2008) How I treat cobalamin (vitamin B12) deficiency. Blood 112, 22142221.CrossRefGoogle Scholar
Carmel, R (2008) Efficacy and safety of fortification and supplementation with vitamin B12: biochemical and physiological effects. Food Nutr Bull 29, S177S187.CrossRefGoogle ScholarPubMed
Thakkar, K, Billa, G (2015) Treatment of vitamin B12 deficiency–Methylcobalamine? Cyancobalamine? Hydroxocobalamin?—clearing the confusion. Eur J Clin Nutr 69, 12.CrossRefGoogle ScholarPubMed
Wolffenbuttel, BH, Wouters, HJ, Heiner-Fokkema, MR, van der Klauw, MM (2019) The many faces of cobalamin (vitamin B12) deficiency. Mayo Clin Proc Innov Qual Outcomes 3, 200214.CrossRefGoogle Scholar
Das, JK, Salam, RA, Mahmood, SB et al. (2019) Food fortification with multiple micronutrients: impact on health outcomes in general population. Cochrane Database Syst Rev.CrossRefGoogle Scholar
Bird, JK, Barron, R, Pigat, S, Bruins, MJ (2022) Contribution of base diet, voluntary fortified foods and supplements to micronutrient intakes in the UK. J Nutr Sci 11, e51.CrossRefGoogle ScholarPubMed
Hopkins, SM, Gibney, MJ, Nugent, AP et al. (2015) Impact of voluntary fortification and supplement use on dietary intakes and biomarker status of folate and vitamin B-12 in Irish adults. Am J Clin Nutr 101, 11631172.CrossRefGoogle ScholarPubMed
WHO (2009) Recommendations on wheat and maize flour fortification meeting report: Interim consensus statement. World Health Organization.Google Scholar
Paul, C, Brady, DM (2017) Comparative bioavailability and utilization of particular forms of B12 supplements with potential to mitigate B12-related genetic polymorphisms. Integr Med (Encinitas) 16, 42.Google Scholar
MacFarlane, AJ, Shi, Y, Greene-Finestone, LS (2014) High-dose compared with low-dose vitamin B-12 supplement use is not associated with higher vitamin B-12 status in children, adolescents, and older adults. J Nutr 144, 915920.CrossRefGoogle Scholar
de Benoist, B (2008) Conclusions of a WHO Technical Consultation on folate and vitamin B12 deficiencies. Food Nutr Bull 29, S238S244.CrossRefGoogle ScholarPubMed
Woodward, RB (1973) The total synthesis of vitamin B12. Pure Appl Chem 33, 145178.CrossRefGoogle Scholar
Eschenmoser, A (2015) Corrin syntheses. Part I: introduction and overview. Helv Chim Acta 98, 14831600.CrossRefGoogle Scholar
Eschenmoser, A (2015) Introductory remarks on the publication series ‘Corrin syntheses–parts I–VI’. Helv Chim Acta 98, 14751482.CrossRefGoogle Scholar
Eschenmoser, A, Wintner, CE (1977) Natural product synthesis and vitamin B12: total synthesis of vitamin B12 provided a framework for exploration in several areas of organic chemistry. Science 196, 14101420.CrossRefGoogle Scholar
Riether, D, Mulzer, J (2003) Total synthesis of cobyric acid: historical development and recent synthetic innovations. European J Org Chem 2003, 3045.3.0.CO;2-I>CrossRefGoogle Scholar
Acevedo-Rocha, CG, Gronenberg, LS, Mack, M et al. (2019) Microbial cell factories for the sustainable manufacturing of B vitamins. Curr Opin Biotechnol 56, 1829.CrossRefGoogle ScholarPubMed
Wang, Y, Liu, L, Jin, Z, Zhang, D (2021) Microbial cell factories for green production of vitamins. Front Bioeng Biotechnol 9, 661562.CrossRefGoogle ScholarPubMed
Ledesma-Amaro, RS, Jiménez, A, Revuelta, JL (2013) Microbial production of vitamins. In Microbial production of food ingredients, enzymes and nutraceuticals, pp. 571594 [McNeil, B, Archer, D, Giavasis, I and Harvey, L, editors] Sawston, United Kingtom: Woodhead Publishing.CrossRefGoogle Scholar
Shimizu, S (2001) Vitamins and Related Compounds: Microbial Production. In Biotechnology Set, pp. 318–340.CrossRefGoogle Scholar
Meyer Zu Berstenhorst, S, Hohmann, H, Stahmann, K (2009) Vitamins and vitamin-like compounds: microbial production. In Encyclopedia of Microbiology, 3rd ed., pp. 549561 [Schaechter, M, editor]. Amsterdam, Netherlands: Elsevier.CrossRefGoogle Scholar
Laudert, D, Hohmann, HP (2011) Application of Enzymes and Microbes for the Industrial Production of Vitamins and Vitamin-Like Compounds. In Comprehensive Biotechnology, pp. 583602 [Moo-Young, M, editor]. Burlington: Academic Press.CrossRefGoogle Scholar
de Carvalho, CC (2017) Whole cell biocatalysts: essential workers from Nature to the industry. Microb Biotechnol 10, 250263.CrossRefGoogle ScholarPubMed
Binod, P, Sindhu, R, Pandey, A (2010) Production of vitamins. In Comprehensive Food Fermentation and Biotechnology.Google Scholar
Survase, SA, Bajaj, IB, Singhal, RS (2006) Biotechnological Production of Vitamins. Food Technol Biotechnol 44, 381396.Google Scholar
Burgess, CM, Smid, EJ, van Sinderen, D (2009) Bacterial vitamin B2, B11 and B12 overproduction: an overview. Int J Food Microbiol 133, 17.CrossRefGoogle ScholarPubMed
Nguyen-Vo, TP, Ainala, SK, Kim, J-R, Park, S (2018) Analysis and characterization of coenzyme B12 biosynthetic gene clusters and improvement of B12 biosynthesis in Pseudomonas denitrificans ATCC 13867. FEMS Microbiol Lett 365, fny211.CrossRefGoogle ScholarPubMed
de Assis, DA, Matte, C, Aschidamini, B et al. (2020) Biosynthesis of vitamin B12 by Propionibacterium freudenreichii subsp. shermanii ATCC 13673 using liquid acid protein residue of soybean as culture medium. Biotechnol Prog 36, e3011.CrossRefGoogle Scholar
Li Kt, Peng Wf, Zhou, J et al. (2013) Establishment of beet molasses as the fermentation substrate for industrial vitamin B12 production by Pseudomonas denitrificans . J Chem Technol Biotechnol 88, 17301735.Google Scholar
Zhang, Y, Liu, J-Z, Huang, J-S, Mao, Z-W (2010) Genome shuffling of Propionibacterium shermanii for improving vitamin B12 production and comparative proteome analysis. J Biotechnol 148, 139143.CrossRefGoogle ScholarPubMed
Li, K-T, Liu, D-H, Li, Y-L et al. (2008) Improved large-scale production of vitamin B12 by Pseudomonas denitrificans with betaine feeding. Bioresour Technol 99, 85168520.CrossRefGoogle ScholarPubMed
Li, K-T, Liu, D-H, Zhuang, Y-P et al. (2008) Influence of Zn2+, Co2+ and dimethylbenzimidazole on vitamin B12 biosynthesis by Pseudomonas denitrificans . World J Microbiol Biotechnol 24, 25252530.CrossRefGoogle Scholar
Li, KT, Zhou, J, Cheng, X, Sj, Wei (2012) Study on the dissolved oxygen control strategy in large-scale vitamin B12 fermentation by Pseudomonas denitrificans . J Chem Technol Biotechnol 87, 16481653.CrossRefGoogle Scholar
Wang, P, Wang, Y, Su, Z (2012) Improvement of adenosylcobalamin production by metabolic control strategy in Propionibacterium freudenreichii . Appl Biochem Biotechnol 167, 6272.CrossRefGoogle ScholarPubMed
Wang, P, Zhang, Z, Jiao, Y et al. (2015) Improved propionic acid and 5, 6-dimethylbenzimidazole control strategy for vitamin B12 fermentation by Propionibacterium freudenreichii . J Biotechnol 193, 123129.CrossRefGoogle ScholarPubMed
Wang, Z-J, Shi, H-L, Wang, P (2016) The online morphology control and dynamic studies on improving vitamin B 12 production by Pseudomonas denitrificans with online capacitance and specific oxygen consumption rate. Appl Biochem Biotechnol 179, 11151127.CrossRefGoogle ScholarPubMed
Xia, W, Chen, W, Peng, W-F, Li, K-T (2015) Industrial vitamin B 12 production by Pseudomonas denitrificans using maltose syrup and corn steep liquor as the cost-effective fermentation substrates. Bioprocess Biosyst Eng 38, 10651073.CrossRefGoogle ScholarPubMed
Liu, J, Liu, Y, Wu, J et al. (2021) Metabolic profiling analysis of the vitamin B12 producer Propionibacterium freudenreichii . Microbiologyopen 10, e1199.CrossRefGoogle ScholarPubMed
Gardner, N, Champagne, C (2005) Production of Propionibacterium shermanii biomass and vitamin B12 on spent media. J Appl Microbiol 99, 12361245.CrossRefGoogle ScholarPubMed
Piao, Y, Yamashita, M, Kawaraichi, N et al. (2004) Production of vitamin B12 in genetically engineered Propionibacterium freudenreichii . J Biosci Bioeng 98, 167173.CrossRefGoogle ScholarPubMed
Kang, Z, Zhang, J, Zhou, J et al. (2012) Recent advances in microbial production of δ-aminolevulinic acid and vitamin B12. Biotechnol Adv 30, 15331542.CrossRefGoogle ScholarPubMed
Piwowarek, K, Lipińska, E, Hać-Szymańczuk, E et al. (2018) Research on the ability of propionic acid and vitamin B12 biosynthesis by Propionibacterium freudenreichii strain T82. Antonie Van Leeuwenhoek 111, 921932.CrossRefGoogle ScholarPubMed
Piwowarek, K, Lipińska, E, Hać-Szymańczuk, E et al. (2018) Propionibacterium spp.—source of propionic acid, vitamin B12, and other metabolites important for the industry. Appl Microbiol Biotechnol 102, 515538.CrossRefGoogle ScholarPubMed
Peng, W, Cheng, X, Zhang Hy, Li KT (2014) The metabolic characteristicsof high-production vitamin B12 by Pseudomonas denitrificans under dissolved oxygen step-wise reduction. J Chem Technol Biotechnol 89, 13961401.CrossRefGoogle Scholar
Kośmider, A, Białas, W, Kubiak, P et al. (2012) Vitamin B12 production from crude glycerol by Propionibacterium freudenreichii ssp. shermanii: optimization of medium composition through statistical experimental designs. Bioresour Technol 105, 128133.CrossRefGoogle ScholarPubMed
Hajfarajollah, H, Mokhtarani, B, Mortaheb, H, Afaghi, A (2015) Vitamin B 12 biosynthesis over waste frying sunflower oil as a cost effective and renewable substrate. J Food Sci Technol 52, 32733282.Google Scholar
Dank, A, Biel, G, Abee, T, Smid, EJ (2022) Microaerobic metabolism of lactate and propionate enhances vitamin B12 production in Propionibacterium freudenreichii . Microb Cell Fact 21, 225.CrossRefGoogle ScholarPubMed
Kumar, R, Singh, U, Tiwari, A et al. (2023) Vitamin B12: strategies for enhanced production, fortified functional food products and health benefits. Process Biochem 127, 4455.CrossRefGoogle Scholar
Mani, I (2020) Microbial Production of Vitamins. In Engineering of Microbial Biosynthetic Pathways, pp. 143-152 [AKS Vijai Singh, Poonam Bhargava, Madhvi Joshi, Chaitanya G. Joshi, editor].CrossRefGoogle Scholar
Fang, H, Kang, J, Zhang, D (2017) Microbial production of vitamin B12: a review and future perspectives. Microb Cell Fact 16, 114.CrossRefGoogle ScholarPubMed
Fang, H, Li, D, Kang, J et al. (2018) Metabolic engineering of Escherichia coli for de novo biosynthesis of vitamin B12. Nat Commun 9, 112.CrossRefGoogle Scholar
Dong, H, Li, S, Fang, H et al. (2016) A newly isolated and identified vitamin B12 producing strain: Sinorhizobium meliloti 320. Bioprocess Biosyst Eng 39, 15271537.CrossRefGoogle ScholarPubMed
Noh, MH, Lim, HG, Moon, D et al. (2020) Auxotrophic selection strategy for improved production of coenzyme B12 in Escherichia coli . iScience 23, 100890.CrossRefGoogle Scholar
Ko, Y, Ashok, S, Ainala, SK et al. (2014) Coenzyme B12 can be produced by engineered Escherichia coli under both anaerobic and aerobic conditions. Biotechnol J 9, 15261535.CrossRefGoogle ScholarPubMed
Mohammed, Y, Lee, B, Kang, Z, Du, G (2014) Development of a two-step cultivation strategy for the production of vitamin B12 by Bacillus megaterium . Microb Cell Fact 13, 110.CrossRefGoogle ScholarPubMed
Cai, Y, Xia, M, Dong, H et al. (2018) Engineering a vitamin B12 high-throughput screening system by riboswitch sensor in Sinorhizobium meliloti . BMC Biotechnol 18, 111.CrossRefGoogle ScholarPubMed
Zhao, T, Cheng, K, Yh, Cao et al. (2019) Identification and antibiotic resistance assessment of Ensifer adhaerens YX1, a vitamin B12-producing strain used as a food and feed additive. J Food Sci 84, 29252931.CrossRefGoogle Scholar
Vu, HT, Itoh, H, Ishii, S et al. (2013) Identification and phylogenetic characterization of cobalamin biosynthetic genes of Ensifer adhaerens . Microbes Environ 28, 153155.Google Scholar
Liu, Z, Dong, H, Wu, X et al. (2021) Identification of a xylose-inducible promoter and its application for improving vitamin B12 production in Sinorhizobium meliloti . Biotechnol Appl Biochem 68, 856864.CrossRefGoogle ScholarPubMed
Li, D, Fang, H, Gai, Y et al. (2020) Metabolic engineering and optimization of the fermentation medium for vitamin B 12 production in Escherichia coli . Bioprocess Biosyst Eng 43, 17351745.CrossRefGoogle ScholarPubMed
Biedendieck, R, Malten, M, Barg, H et al. (2010) Metabolic engineering of cobalamin (vitamin B12) production in Bacillus megaterium . Microb Biotechnol 3, 2437.CrossRefGoogle ScholarPubMed
Moore, SJ, Mayer, MJ, Biedendieck, R et al. (2014) Towards a cell factory for vitamin B12 production in Bacillus megaterium: bypassing of the cobalamin riboswitch control elements. N Biotechnol 31, 553561.CrossRefGoogle ScholarPubMed
Xu, S, Xiao, Z, Yu, S et al. (2022) Enhanced cobalamin biosynthesis in Ensifer adhaerens by regulation of key genes with gradient promoters. Synth Syst Biotechnol 7, 941948.CrossRefGoogle ScholarPubMed
Malik, KA, Maqbool, A (2020) Transgenic crops for biofortification. Front Sustain Food Syst 4, 571402.CrossRefGoogle Scholar
Bouis, HE, Saltzman, A (2017) Improving nutrition through biofortification: a review of evidence from HarvestPlus, 2003 through 2016. Glob Food Sec 12, 4958.CrossRefGoogle ScholarPubMed
Garg, M, Sharma, N, Sharma, S et al. (2018) Biofortified crops generated by breeding, agronomy, and transgenic approaches are improving lives of millions of people around the world. Front Nutr 5, 12.CrossRefGoogle ScholarPubMed
Titcomb, TJ, Tanumihardjo, SA (2019) Global concerns with B vitamin statuses: biofortification, fortification, hidden hunger, interactions, and toxicity. Compr Rev Food Sci Food Saf 18, 19681984.CrossRefGoogle ScholarPubMed
Oh, S, Cave, G, Lu, C (2021) Vitamin B12 (cobalamin) and micronutrient fortification in food crops using nanoparticle technology. Front Plant Sci 12, 1451.CrossRefGoogle ScholarPubMed
Garg, M, Sharma, A, Vats, S et al. (2021) Vitamins in cereals: a critical review of content, health effects, processing losses, bioaccessibility, fortification, and biofortification strategies for their improvement. Front Nutr 8, 586815.CrossRefGoogle ScholarPubMed
Mozafar, A (1994) Enrichment of some B-vitamins in plants with application of organic fertilizers. Plant Soil 167, 305311.CrossRefGoogle Scholar
Lima, S, Webb, CL, Deery, E et al. (2018) Human intrinsic factor expression for bioavailable vitamin B12 enrichment in microalgae. Biology 7, 19.CrossRefGoogle ScholarPubMed
Rutten, MJ, Bouwman, AC, Sprong, RC et al. (2013) Genetic variation in vitamin B-12 content of bovine milk and its association with SNP along the bovine genome. PLoS One 8, e62382.CrossRefGoogle ScholarPubMed
Duplessis, M, Pellerin, D, Robichaud, R et al. (2019) Impact of diet management and composition on vitamin B12 concentration in milk of Holstein cows. Animal 13, 21012109.CrossRefGoogle ScholarPubMed
Gebreyesus, G, Poulsen, NA, Larsen, MK et al. (2021) Vitamin B12 and transcobalamin in bovine milk: genetic variation and genome-wide association with loci along the genome. JDS Commun 2, 127131.CrossRefGoogle ScholarPubMed
Kincaid, R, Socha, M (2007) Effect of cobalt supplementation during late gestation and early lactation on milk and serum measures. J Dairy Sci 90, 18801886.CrossRefGoogle ScholarPubMed
Akins, MS, Bertics, S, Socha, M, Shaver, R (2013) Effects of cobalt supplementation and vitamin B12 injections on lactation performance and metabolism of Holstein dairy cows. J Dairy Sci 96, 17551768.CrossRefGoogle ScholarPubMed
Stemme, K, Lebzien, P, Flachowsky, G, Scholz, H (2008) The influence of an increased cobalt supply on ruminal parameters and microbial vitamin B12 synthesis in the rumen of dairy cows. Arch Anim Nutr 62, 207218.CrossRefGoogle ScholarPubMed
Bernhardt, C, Zhu, X, Schütz, D et al. (2019) Cobalamin is produced by Acetobacter pasteurianus DSM 3509. Appl Microbiol Biotechnol 103, 38753885.CrossRefGoogle ScholarPubMed
Chamlagain, B, Deptula, P, Edelmann, M et al. (2016) Effect of the lower ligand precursors on vitamin B12 production by food-grade Propionibacteria. Lwt-Food Sci Technol 72, 117124.CrossRefGoogle Scholar
Chamlagain, B, Edelmann, M, Kariluoto, S et al. (2015) Ultra-high performance liquid chromatographic and mass spectrometric analysis of active vitamin B12 in cells of Propionibacterium and fermented cereal matrices. Food Chem 166, 630638.CrossRefGoogle ScholarPubMed
Chamlagain, B, Peltonen, L, Edelmann, M et al. (2021) Bioaccessibility of vitamin B12 synthesized by Propionibacterium freudenreichii and from products made with fermented wheat bran extract. Curr Res Food Sci 4, 499502.CrossRefGoogle ScholarPubMed
Deptula, P, Chamlagain, B, Edelmann, M et al. (2017) Food-like growth conditions support production of active vitamin B12 by Propionibacterium freudenreichii 2067 without DMBI, the lower ligand base, or cobalt supplementation. Front Microbiol 8, 368.CrossRefGoogle ScholarPubMed
Tangyu, M, Fritz, M, Ye, L et al. (2022) Co-cultures of Propionibacterium freudenreichii and Bacillus amyloliquefaciens cooperatively upgrade sunflower seed milk to high levels of vitamin B12 and multiple co-benefits. Microb Cell Fact 21, 123.CrossRefGoogle ScholarPubMed
Xie, C, Coda, R, Chamlagain, B et al. (2018) In situ fortification of vitamin B12 in wheat flour and wheat bran by fermentation with Propionibacterium freudenreichii . J Cereal Sci 81, 133139.CrossRefGoogle Scholar
Xie, C, Coda, R, Chamlagain, B et al. (2021) Fermentation of cereal, pseudo-cereal and legume materials with Propionibacterium freudenreichii and Levilactobacillus brevis for vitamin B12 fortification. Lwt-Food Sci Technol 137, 110431.CrossRefGoogle Scholar
Xie, C, Coda, R, Chamlagain, B et al. (2019) Co-fermentation of Propionibacterium freudenreichii and Lactobacillus brevis in wheat bran for in situ production of vitamin B12. Front Microbiol 10, 1541.CrossRefGoogle ScholarPubMed
Signorini, C, Carpen, A, Coletto, L et al. (2018) Enhanced vitamin B12 production in an innovative lupin tempeh is due to synergic effects of Rhizopus and Propionibacterium in cofermentation. Int J Food Sci Nutr 69, 451457.CrossRefGoogle Scholar
de Assis, DA, Machado, C, Matte, C, Ayub, MAZ (2022) High cell density culture of dairy Propionibacterium sp. and Acidipropionibacterium sp.: a review for food industry applications. Food Bioproc Tech 15, 734749.CrossRefGoogle Scholar
Wolkers–Rooijackers, JC, Endika, MF, Smid, EJ (2018) Enhancing vitamin B12 in lupin tempeh by in situ fortification. Lwt-Food Sci Technol 96, 513518.CrossRefGoogle Scholar
Shi, L, Xu, Y, Zhan, L et al. (2018) Enhancing vitamin B12 content in co-fermented soy-milk via a Lotka Volterra model. Turk J Biochem 43, 671678.CrossRefGoogle Scholar
Hugenschmidt, S, Schwenninger, SM, Gnehm, N, Lacroix, C (2010) Screening of a natural biodiversity of lactic and propionic acid bacteria for folate and vitamin B12 production in supplemented whey permeate. Int Dairy J 20, 852857.CrossRefGoogle Scholar
Hugenschmidt, S, Schwenninger, SM, Lacroix, C (2011) Concurrent high production of natural folate and vitamin B12 using a co-culture process with Lactobacillus plantarum SM39 and Propionibacterium freudenreichii DF13. Process Biochem 46, 10631070.CrossRefGoogle Scholar
Capozzi, V, Russo, P, Duenas, MT et al. (2012) Lactic acid bacteria producing B-group vitamins: a great potential for functional cereals products. Appl Microbiol Biotechnol 96, 13831394.CrossRefGoogle ScholarPubMed
Kittaka-Katsura, H, Ebara, S, Watanabe, F, Nakano, Y (2004) Characterization of corrinoid compounds from a Japanese black tea (Batabata-cha) fermented by bacteria. J Agric Food Chem 52, 909911.CrossRefGoogle ScholarPubMed
Chamlagain, B, Sugito, TA, Deptula, P et al. (2018) In situ production of active vitamin B12 in cereal matrices using Propionibacterium freudenreichii . Food Sci Nutr 6, 6776.CrossRefGoogle Scholar
Coelho, RMD, de Almeida, AL, do Amaral, RQG et al. (2020) Kombucha. Int J Gastron Food Sci 22, 100272.CrossRefGoogle Scholar
Gu, Q, Zhang, C, Song, D et al. (2015) Enhancing vitamin B12 content in soy-yogurt by Lactobacillus reuteri . Int J Food Microbiol 206, 5659.CrossRefGoogle ScholarPubMed
Li, P, Gu, Q, Yang, L et al. (2017) Characterization of extracellular vitamin B12 producing Lactobacillus plantarum strains and assessment of the probiotic potentials. Food Chem 234, 494501.CrossRefGoogle ScholarPubMed
Thompson, HO, Önning, G, Holmgren, K et al. (2020) Fermentation of cauliflower and white beans with Lactobacillus plantarum – impact on levels of riboflavin, folate, vitamin B 12, and amino acid composition. Plant Foods Hum Nutr 75, 236242.CrossRefGoogle ScholarPubMed
Molina, V, Médici, M, de Valdez, GF, Taranto, MP (2012) Soybean-based functional food with vitamin B12-producing lactic acid bacteria. J Funct Foods 4, 831836.CrossRefGoogle Scholar
Bhushan, B, Tomar, S, Chauhan, A (2017) Techno-functional differentiation of two vitamin B12 producing Lactobacillus plantarum strains: an elucidation for diverse future use. Appl Microbiol Biotechnol 101, 697709.CrossRefGoogle ScholarPubMed
Kumari, M, Bhushan, B, Kokkiligadda, A et al. (2021) Vitamin B12 biofortification of soymilk through optimized fermentation with extracellular B12 producing Lactobacillus isolates of human fecal origin. Curr Res Food Sci 4, 646654.CrossRefGoogle ScholarPubMed
Taranto, MP, Vera, JL, Hugenholtz, J et al. (2003) Lactobacillus reuteri CRL1098 produces cobalamin. J Bacteriol 185, 56435647.CrossRefGoogle ScholarPubMed
Torres, AC, Vannini, V, Font, G et al. (2018) Novel pathway for corrinoid compounds production in Lactobacillus . Front Microbiol 9, 2256.CrossRefGoogle ScholarPubMed
Santos, F (2008) Vitamin B 12 synthesis in Lactobacillus reuteri. Wageningen University and Research.Google Scholar
Santos, F, Vera, JL, Lamosa, P et al. (2007) Pseudovitamin is the corrinoid produced by Lactobacillus reuteri CRL1098 under anaerobic conditions. FEBS Lett 581, 48654870.CrossRefGoogle ScholarPubMed
Basavanna, G, Prapulla, SG (2013) Evaluation of functional aspects of Lactobacillus fermentum CFR 2195 isolated from breast fed healthy infants’ fecal matter. J Food Sci Technol 50, 360366.CrossRefGoogle ScholarPubMed
Martín, R, Olivares, M, Marín, M et al. (2005) Characterization of a reuterin-producing Lactobacillus coryniformis strain isolated from a goat’s milk cheese. Int J Food Microbiol 104, 267277.CrossRefGoogle ScholarPubMed
De Angelis, M, Bottacini, F, Fosso, B et al. (2014) Lactobacillus rossiae, a vitamin B12 producer, represents a metabolically versatile species within the genus Lactobacillus . PLoS One 9, e107232.CrossRefGoogle ScholarPubMed
Australian Government Department of Health and Aging NH and MRC (2005) Nutrient Reference Values for Australia and New Zealand Including Recommended Dietary Intakes. Canberra, Australia: National Health and Medical Research Council.Google Scholar
Yates, AA (2001) National nutrition and public health policies: issues related to bioavailability of nutrients when developing dietary reference intakes. J Nutr 131, 1331S1334S.CrossRefGoogle Scholar
Kozyraki, R, Cases, O (2013) Vitamin B12 absorption: mammalian physiology and acquired and inherited disorders. Biochimie 95, 10021007.CrossRefGoogle ScholarPubMed
Berlin, H, Berlin, R, Brante, G (1968) Oral treatment of pernicious anemia with high doses of vitamin B12 without intrinsic factor. Acta Med Scand 184, 247258.CrossRefGoogle ScholarPubMed
Fedosov, SN (2012) Physiological and Molecular Aspects of Cobalamin Transport. In Water Soluble Vitamins: Clinical Research and Future Application, pp. 347367 [Stanger, O, editor]. Dordrecht: Springer Netherlands.CrossRefGoogle Scholar
Shipton, MJ, Thachil, J (2015) Vitamin B12 deficiency – A 21st century perspective. Clin Med (Lond) 15, 145150.CrossRefGoogle ScholarPubMed
Quadros, EV, Nakayama, Y, Sequeira, JM (2009) The protein and the gene encoding the receptor for the cellular uptake of transcobalamin-bound cobalamin. Blood 113, 186192.CrossRefGoogle Scholar
Watkins, D, Rosenblatt, DS (2022) Inherited defects of cobalamin metabolism. Vitam Horm 119, 355376.CrossRefGoogle ScholarPubMed
Bloomfield, FJ, Scott, JM (1972) Identification of a new vitamin B 12 binder (transcobalamin 3) in normal human serum. Br J Haematol 22, 3342.CrossRefGoogle ScholarPubMed
Burger, RL, Schneider, RJ, Mehlman, CS, Allen, RH (1975 ) Human plasma R-type vitamin B12-binding proteins. II. The role of transcobalamin I, transcobalamin III, and the normal granulocyte vitamin B12-binding protein in the plasma transport of vitamin B12. J Biol Chem 250, 77077713.CrossRefGoogle ScholarPubMed
Fràter-Schröder, M, Hitzig, WH, Bütler, R (1979) Studies on transcobalamin (TC). 1. Detection of TC II isoproteins in human serum. Blood 53, 193203.CrossRefGoogle ScholarPubMed
Meyer, LM, Miller, IF, Gizis, E et al. (1974) Delivery of vitamin B12 to human lymphocytes by transcobalamins I, II and 3. Proc Soc Exp Biol Med 146, 747750.CrossRefGoogle ScholarPubMed
Wickramasinghe, SN, England, JM, Saunders, JE, Down, MC (1975) Role of transcobalamins I, II, and III in the transfer of vitamin B12 to human bone marrow cells in vitro . Acta Haematol 54, 8994.CrossRefGoogle ScholarPubMed
Herrmann, W, Obeid, R (2012) Cobalamin deficiency. Subcell Biochem 56, 301322.CrossRefGoogle ScholarPubMed
Manzanares, W, Hardy, G (2010) Vitamin B12: the forgotten micronutrient for critical care. Curr Opin Clin Nutr Metab Care 13, 662668.CrossRefGoogle ScholarPubMed
Kräutler, B (2012) Biochemistry of B12-cofactors in human metabolism. Subcell Biochem 56, 323346.CrossRefGoogle ScholarPubMed
Mascarenhas, R, Gouda, H, Ruetz, M, Banerjee, R (2022) Human B12-dependent enzymes: methionine synthase and methylmalonyl-CoA mutase. Meth Enzymol 668, 309326.CrossRefGoogle Scholar
Watkins, D, Ru, M, Hwang, HY et al. (2002) Hyperhomocysteinemia due to methionine synthase deficiency, cblG: structure of the MTR gene, genotype diversity, and recognition of a common mutation, P1173L. Am J Hum Genet 71, 143153.CrossRefGoogle ScholarPubMed
Kim, J, Kim, H, Roh, H, Kwon, Y (2018) Causes of hyperhomocysteinemia and its pathological significance. Arch Pharm Res 41, 372383.CrossRefGoogle ScholarPubMed
McCaddon, A, Miller, JW (2023) Homocysteine—a retrospective and prospective appraisal. Front Nutr 10, 1179807.CrossRefGoogle ScholarPubMed
Allen, LH (2012) Vitamin B-12. Adv Nutr 3, 5455.CrossRefGoogle ScholarPubMed
Harrington, DJ (2017) Laboratory assessment of vitamin B12 status. J Clin Pathol 70, 168173.CrossRefGoogle ScholarPubMed
Fedosov, SN (2010) Metabolic signs of vitamin B(12) deficiency in humans: computational model and its implications for diagnostics. Metabolism 59, 11241138.CrossRefGoogle ScholarPubMed
Fedosov, SN, Brito, A, Miller, JW et al. (2015) Combined indicator of vitamin B12 status: modification for missing biomarkers and folate status and recommendations for revised cut-points. Clin Chem Lab Med 53, 12151225.CrossRefGoogle ScholarPubMed
Carmel, R (2007) The disappearance of cobalamin absorption testing: a critical diagnostic loss. J Nutr 137, 24812484.CrossRefGoogle ScholarPubMed
von Castel-Roberts, KM, Morkbak, AL, Nexo, E et al. (2007) Holo-transcobalamin is an indicator of vitamin B-12 absorption in healthy adults with adequate vitamin B-12 status. Am J Clin Nutr 85, 10571061.CrossRefGoogle ScholarPubMed
Hvas, A-M, Morkbak, AL, Nexo, E (2007) Plasma holotranscobalamin compared with plasma cobalamins for assessment of vitamin B12 absorption; optimisation of a non-radioactive vitamin B12 absorption test (CobaSorb). Clin Chim Acta 376, 150154.CrossRefGoogle ScholarPubMed
Hardlei, TF, Mørkbak, AL, Bor, MV et al. (2010) Assessment of vitamin B12 absorption based on the accumulation of orally administered cyanocobalamin on transcobalamin. Clin Chem 56, 432436.CrossRefGoogle ScholarPubMed
Nexo, E, Hoffmann-Lücke, E (2011) Holotranscobalamin, a marker of vitamin B-12 status: analytical aspects and clinical utility. Am J Clin Nutr 94, 359S365S.CrossRefGoogle ScholarPubMed
Alonso Nr, Granada ML, Salinas, I et al. (2005) Serum pepsinogen I: an early marker of pernicious anemia in patients with type 1 diabetes. J Clin Endocrinol Metab 90, 52545258.CrossRefGoogle Scholar
Herbert, V (1994) Staging vitamin B-12 (cobalamin) status in vegetarians. Am J Clin Nutr 59, 1213S1222S.CrossRefGoogle ScholarPubMed
Carmel, R (2013) Diagnosis and management of clinical and subclinical cobalamin deficiencies: why controversies persist in the age of sensitive metabolic testing. Biochimie 95, 10471055.CrossRefGoogle ScholarPubMed
Higginbottom, MC, Sweetman, L, Nyhan, WL (1978) A syndrome of methylmalonic aciduria, homocystinuria, megaloblastic anemia and neurologic abnormalities in a vitamin B12-deficient breast-fed infant of a strict vegetarian. N Engl J Med 299, 317323.CrossRefGoogle Scholar
Monsen, A-LB, Ueland, PM, Vollset, SE et al. (2001) Determinants of cobalamin status in newborns. Pediatrics 108, 624.CrossRefGoogle Scholar
Berenson, AB, Rahman, M (2012) Effect of hormonal contraceptives on vitamin B12 level and the association of the latter with bone mineral density. Contraception 86, 481487.CrossRefGoogle ScholarPubMed
Tal, S, Shavit, Y, Stern, F, Malnick, S (2010) Association between vitamin B12 levels and mortality in hospitalized older adults. J Am Geriatr Soc 58, 523526.CrossRefGoogle ScholarPubMed
Chanarin, I (1980) Cobalamins and nitrous oxide: a review. J Clin Pathol 33, 909916.CrossRefGoogle ScholarPubMed
Rosenblatt, DS, Cooper, BA (1987) Inherited disorders of vitamin B12 metabolism. Blood Rev 1, 177182.CrossRefGoogle ScholarPubMed
Dali-Youcef, N, Andrès, E (2008) An update on cobalamin deficiency in adults. QJM Int J Med 102, 1728.CrossRefGoogle ScholarPubMed
Lachner, C, Steinle, NI, Regenold, WT (2012) The neuropsychiatry of vitamin B12 deficiency in elderly patients. J Neuropsychiatry Clin Neurosci 24, 515.CrossRefGoogle ScholarPubMed
Reynolds, E (2006) Vitamin B12, folic acid, and the nervous system. Lancet Neurol 5, 949960.CrossRefGoogle ScholarPubMed
McCaddon, A (2013) Vitamin B12 in neurology and ageing; Clinical and genetic aspects. Biochimie 95, 10661076.CrossRefGoogle ScholarPubMed
Hutto, BR (1997) Folate and cobalamin in psychiatric illness. Compr Psychiatry 38, 305314.CrossRefGoogle ScholarPubMed
Obeid, R, McCaddon, A, Herrmann, W (2007) The role of hyperhomocysteinemia and B-vitamin deficiency in neurological and psychiatric diseases. Clin Chem Lab Med 45, 15901606.CrossRefGoogle ScholarPubMed
Norbert Goebels, M.D., Michael Soyka, M.D. (2000) Dementia associated with vitamin B12 deficiency. J Neuropsychiatry Clin Neurosci 12, 389394.CrossRefGoogle Scholar
Metz, J (1992) Cobalamin deficiency and the pathogenesis of nervous system disease. Annu Rev Nutr 12, 5979.CrossRefGoogle ScholarPubMed
Finkelstein, JL, Layden, AJ, Stover, PJ (2015) Vitamin B-12 and perinatal health. Adv Nutr 6, 552563.CrossRefGoogle ScholarPubMed
Jiang, T, Christian, P, Khatry, SK et al. (2005) Micronutrient deficiencies in early pregnancy are common, concurrent, and vary by season among rural Nepali pregnant women. J Nutr 135, 11061112.CrossRefGoogle ScholarPubMed
Allen, LH, Rosenberg, IH, Oakley, GP, Omenn, GS (2010) Considering the case for vitamin B12 fortification of flour. Food Nutr Bull 31, S36S46.CrossRefGoogle ScholarPubMed
Thankachan, P, Rah, JH, Thomas, T et al. (2012) Multiple micronutrient-fortified rice affects physical performance and plasma vitamin B-12 and homocysteine concentrations of Indian school children. J Nutr 142, 846852.CrossRefGoogle ScholarPubMed
Allen, LH (2009) How common is vitamin B-12 deficiency? Am J Clin Nutr 89, 693S696S.CrossRefGoogle ScholarPubMed
Refsum, H, Smith, AD (2008) Are we ready for mandatory fortification with vitamin B-12?, vol. 88, pp. 253254, Oxford University Press.Google ScholarPubMed
Dhonukshe-Rutten, RA, van Zutphen, M, de Groot, LC et al. (2005) Effect of supplementation with cobalamin carried either by a milk product or a capsule in mildly cobalamin-deficient elderly Dutch persons–. Am J Clin Nutr 82, 568574.CrossRefGoogle ScholarPubMed
McNulty, H, Scott, JM (2008) Intake and status of folate and related B-vitamins: considerations and challenges in achieving optimal status. Br J Nutr 99, S48S54.CrossRefGoogle Scholar
McNulty, H, Ward, M, Hoey, L et al. (2019) Addressing optimal folate and related B-vitamin status through the lifecycle: health impacts and challenges. Proc Nutr Soc 78, 449462.CrossRefGoogle ScholarPubMed
Blacher, J, Czernichow, S, Raphaöl, M et al. (2007) Very low oral doses of vitamin B-12 increase serum concentrations in elderly subjects with food-bound vitamin B-12 malabsorption. J Nutr 137, 373378.CrossRefGoogle ScholarPubMed
Porter, K, Hoey, L, Hughes, CF et al. (2016) Causes, consequences and public health implications of low B-vitamin status in ageing. Nutrients 8, 725.CrossRefGoogle ScholarPubMed
Sanchez, H, Albala, C, Lera, L et al. (2013) Effectiveness of the National Program of Complementary Feeding for older adults in Chile on vitamin B12 status in older adults; secondary outcome analysis from the CENEX Study (ISRCTN48153354). Nutr J 12, 18.CrossRefGoogle ScholarPubMed
Eussen, SJ, de Groot, LC, Clarke, R et al. (2005) Oral cyanocobalamin supplementation in older people with vitamin B12 deficiency: a dose-finding trial. Arch Intern Med 165, 11671172.CrossRefGoogle ScholarPubMed
Oakley, G Jr (1997) Let’s increase folic acid fortification and include vitamin B-12, vol. 65, pp. 18891890, Oxford University Press.Google ScholarPubMed
WHO (2018) Guideline: Fortification of rice with vitamins and minerals as a public health strategy. Geneva, Switzerland.Google Scholar
WHO (2022) Guideline: fortification of wheat flour with vitamins and minerals as a public health strategy.Google Scholar
Mills, JL (2000) Fortification of foods with folic acid—how much is enough?, vol. 342, pp. 14421445, Mass Medical Soc. Google Scholar
Mills, JL, Molloy, AM, Reynolds, EH (2018) Do the benefits of folic acid fortification outweigh the risk of masking vitamin B12 deficiency? BMJ 360, k724.CrossRefGoogle Scholar
Selhub, J, Miller, JW, Troen, AM et al. (2022) Perspective: the high-folate–low-vitamin B-12 interaction is a novel cause of vitamin B-12 depletion with a specific etiology—a hypothesis. Adv Nutr 13, 1633.CrossRefGoogle ScholarPubMed
Selhub, J, Morris, MS, Jacques, PF (2007) In vitamin B12 deficiency, higher serum folate is associated with increased total homocysteine and methylmalonic acid concentrations. Proc Natl Acad Sci 104, 1999520000.CrossRefGoogle ScholarPubMed
Selhub, J, Morris, MS, Jacques, PF (2008) Reply to Quinlivan: postfortification, folate intake in vitamin B12 deficiency is positively related to homocysteine and methylmalonic acid. Proc Natl Acad Sci 105, E8E8.CrossRefGoogle Scholar
Selhub, J, Morris, MS, Jacques, PF, Rosenberg, IH (2009) Folate–vitamin B-12 interaction in relation to cognitive impairment, anemia, and biochemical indicators of vitamin B-12 deficiency. Am J Clin Nutr 89, 702S706S.CrossRefGoogle ScholarPubMed
Selhub, J, Paul, L (2011) Folic acid fortification: why not vitamin B12 also? Biofactors 37, 269271.CrossRefGoogle Scholar
Pan American Health Organization (2004) Centers for Disease Control and Prevention Recommended Levels of Folic Acid and Vitamin B12. Food and Nutrition Program of the Pan American Health Organization.Google Scholar
Hirsch, S, de la Maza, P, Barrera, G et al. (2002) The Chilean flour folic acid fortification program reduces serum homocysteine levels and masks vitamin B-12 deficiency in elderly people. J Nutr 132, 289291.CrossRefGoogle ScholarPubMed
Herbert, V, Bigaouette, J (1997) Call for endorsement of a petition to the Food and Drug Administration to always add vitamin B-12 to any folate fortification or supplement. Am J Clin Nutr 65, 572573.CrossRefGoogle ScholarPubMed
Garrod, MG, Buchholz, BA, Miller, JW et al. (2019) Vitamin B12 added as a fortificant to flour retains high bioavailability when baked in bread. Nucl Instrum Methods Phys Res B 438, 136140.CrossRefGoogle ScholarPubMed
Ray, J, Vermeulen, M, Langman, L et al. (2003) Persistence of vitamin B12 insufficiency among elderly women after folic acid food fortification. Clin Biochem 36, 387391.CrossRefGoogle ScholarPubMed
Ray, JG, Cole, DE, Boss, SC (2000) An Ontario-wide study of vitamin B12, serum folate, and red cell folate levels in relation to plasma homocysteine: is a preventable public health issue on the rise? Clin Biochem 33, 337343.CrossRefGoogle ScholarPubMed
Brouwer, I, Verhoef, P (2007) Folic acid fortification: is masking of vitamin B-12 deficiency what we should really worry about?, vol. 86, pp. 897898, Oxford University Press.Google ScholarPubMed
Allen, LH (2012) Pros and cons of increasing folic acid and vitamin B12 intake by fortification. Meeting Micronutrient Requirements for Health and Development 70, 175183.CrossRefGoogle ScholarPubMed
Ding, Z, Luo, L, Guo, S et al. (2022) Non-linear association between folate/vitamin B12 status and cognitive function in older adults. Nutrients 14, 2443.CrossRefGoogle ScholarPubMed
Deng, Y, Wang, D, Wang, K, Kwok, T (2017) High serum folate is associated with brain atrophy in older diabetic people with vitamin B12 deficiency. J Nutr Health Aging 21, 10651071.CrossRefGoogle ScholarPubMed
Czernichow, S, Noisette, N, Blacher, J et al. (2005) Case for folic acid and vitamin B12 fortification in Europe. Semin Vasc Med 5, 156162.CrossRefGoogle ScholarPubMed
Xu, H, Wang, S, Gao, F, Li, C (2022) Vitamin B6, B9, and B12 intakes and cognitive performance in elders: National Health and Nutrition Examination Survey, 2011–2014. Neuropsychiatr Dis Treat 18, 537.CrossRefGoogle ScholarPubMed
Reynolds, E (2016) What is the safe upper intake level of folic acid for the nervous system? Implications for folic acid fortification policies. Eur J Clin Nutr 70, 537540.CrossRefGoogle ScholarPubMed
Reynolds, EH (2017) The risks of folic acid to the nervous system in vitamin B12 deficiency: rediscovered in the era of folic acid fortification policies. J Neurol Neurosurg Psychiatry 88, 10971098.CrossRefGoogle Scholar
Morris, MS, Jacques, PF, Rosenberg, IH, Selhub, J (2010) Circulating unmetabolized folic acid and 5-methyltetrahydrofolate in relation to anemia, macrocytosis, and cognitive test performance in American seniors. Am J Clin Nutr 91, 17331744.CrossRefGoogle ScholarPubMed
Varela-Moreiras, G, Murphy, MM, Scott, JM (2009) Cobalamin, folic acid, and homocysteine. Nutr Rev 67, S69S72.CrossRefGoogle ScholarPubMed
Cuskelly, GJ, Mooney, KM, Young, IS (2007) Folate and vitamin B12: friendly or enemy nutrients for the elderly: symposium on ‘Micronutrients through the life cycle’. Proc Nutr Soc 66, 548558.CrossRefGoogle ScholarPubMed
Johnson, MA (2007) If high folic acid aggravates vitamin B12 deficiency what should be done about it? Nutr Rev 65, 451458.CrossRefGoogle Scholar
Quinlivan, EP (2008) In vitamin B12 deficiency, higher serum folate is associated with increased homocysteine and methylmalonic acid concentrations. Proc Natl Acad Sci 105, E7E7.CrossRefGoogle ScholarPubMed
Doets, EL, Ueland, PM, Tell, GS et al. (2014) Interactions between plasma concentrations of folate and markers of vitamin B12 status with cognitive performance in elderly people not exposed to folic acid fortification: the Hordaland Health Study. Br J Nutr 111, 10851095.CrossRefGoogle Scholar
Wyckoff, KF, Ganji, V (2007) Proportion of individuals with low serum vitamin B-12 concentrations without macrocytosis is higher in the post–folic acid fortification period than in the pre–folic acid fortification period. Am J Clin Nutr 86, 11871192.CrossRefGoogle ScholarPubMed
MacFarlane, AJ, Greene-Finestone, LS, Shi, Y (2011) Vitamin B-12 and homocysteine status in a folate-replete population: results from the Canadian Health Measures Survey. Am J Clin Nutr 94, 10791087.CrossRefGoogle Scholar
Sawaengsri, H, Bergethon, PR, Qiu, WQ et al. (2016) Transcobalamin 776C→ G polymorphism is associated with peripheral neuropathy in elderly individuals with high folate intake. Am J Clin Nutr 104, 16651670.CrossRefGoogle ScholarPubMed
Miller, JW, Garrod, MG, Allen, LH et al. (2009) Metabolic evidence of vitamin B-12 deficiency, including high homocysteine and methylmalonic acid and low holotranscobalamin, is more pronounced in older adults with elevated plasma folate. Am J Clin Nutr 90, 15861592.CrossRefGoogle ScholarPubMed
Berry, RJ (2019) Lack of historical evidence to support folic acid exacerbation of the neuropathy caused by vitamin B12 deficiency. Am J Clin Nutr 110, 554561.CrossRefGoogle ScholarPubMed
Morris, MS, Jacques, PF, Rosenberg, IH, Selhub, J (2007) Folate and vitamin B-12 status in relation to anemia, macrocytosis, and cognitive impairment in older Americans in the age of folic acid fortification. Am J Clin Nutr 85, 193200.CrossRefGoogle ScholarPubMed
Mills, JL, Von Kohorn, I, Conley, MR et al. (2003) Low vitamin B-12 concentrations in patients without anemia: the effect of folic acid fortification of grain. Am J Clin Nutr 77, 14741477.CrossRefGoogle Scholar
Metz, J, McNeil, A, Levin, M (2004) The relationship between serum cobalamin concentration and mean red cell volume at varying concentrations of serum folate. Clin Lab Haematol 26, 323325.CrossRefGoogle ScholarPubMed
Malouf, R, Evans, JG (2008) Folic acid with or without vitamin B12 for the prevention and treatment of healthy elderly and demented people. Cochrane Database Syst Rev.CrossRefGoogle Scholar
Friel, JK (2002) Folate supplements and the masking of vitamin B-12 deficiency. J Nutr 132, 20872087.CrossRefGoogle ScholarPubMed
O’Connor, DM, Laird, EJ, Carey, D et al. (2020) Plasma concentrations of vitamin B12 and folate and global cognitive function in an older population: cross-sectional findings from The Irish Longitudinal Study on Ageing (TILDA). Br J Nutr 124, 602610.CrossRefGoogle Scholar
Carter, B, Zenasni, Z, Moat, SJ et al. (2021) Plasma methylmalonic acid concentration in folic acid–supplemented depressed patients with low or marginal vitamin B-12: a randomized trial. J Nutr 151, 37383745.CrossRefGoogle ScholarPubMed
Qi, YP, Do, AN, Hamner, HC et al. (2014) The prevalence of low serum vitamin B-12 status in the absence of anemia or macrocytosis did not increase among older US adults after mandatory folic acid fortification. J Nutr 144, 170176.CrossRefGoogle Scholar
Crider, KS, Bailey, LB, Berry, RJ (2011) Folic acid food fortification—its history, effect, concerns, and future directions. Nutrients 3, 370384.CrossRefGoogle ScholarPubMed
GFDx (2022) Global Fortification Data Exchange. https://fortificationdata.org/map-number-of-nutrients/ Day Month 2022)Google Scholar
FFI (2022) Food Fortification Initiative. https://www.ffinetwork.org/country-profiles (accessed Day Month 2022)Google Scholar
Engle-Stone, R, Nankap, M, Ndjebayi, AO et al. (2017) Iron, zinc, folate, and vitamin B-12 status increased among women and children in Yaounde and Douala, Cameroon, 1 year after introducing fortified wheat flour. J Nutr 147, 14261436.CrossRefGoogle Scholar
Bobrek, KS, Broersen, B, Aburto, NJ et al. (2021) Most national, mandatory flour fortification standards do not align with international recommendations for iron, zinc, and vitamin B12 levels. Food Policy 99, 101996.CrossRefGoogle Scholar
Muthayya, S, Hall, J, Bagriansky, J et al. (2012) Rice fortification: an emerging opportunity to contribute to the elimination of vitamin and mineral deficiency worldwide. Food Nutr Bull 33, 296307.CrossRefGoogle Scholar
Sirohi, A, Pundhir, A, Ghosh, S (2018) Food fortification: a nutritional management strategy in India. Innovare J Food Sci 6, 18.Google Scholar
FSSAI (2018) Food Safety and Standards (Fortification of Foods) Regulations.Google Scholar
Laird, EJ, O’Halloran, AM, Carey, D et al. (2018) Voluntary fortification is ineffective to maintain the vitamin B12 and folate status of older Irish adults: evidence from the Irish Longitudinal Study on Ageing (TILDA). Br J Nutr 120, 111120.CrossRefGoogle ScholarPubMed
Dunn, ML, Jain, V, Klein, BP (2014) Stability of key micronutrients added to fortified maize flours and corn meal. Ann N Y Acad Sci 1312, 1525.CrossRefGoogle ScholarPubMed
Lu, B, Ren, Y, Huang, B et al. (2008) Simultaneous determination of four water-soluble vitamins in fortified infant foods by ultra-performance liquid chromatography coupled with triple quadrupole mass spectrometry. J Chromatogr Sci 46, 225232.CrossRefGoogle ScholarPubMed
Cardoso, RV, Fernandes, Â, Gonzaléz-Paramás, AM et al. (2019) Flour fortification for nutritional and health improvement: a review. Food Res Int 125, 108576.CrossRefGoogle Scholar
Leyvraz, M, Laillou, A, Rahman, S et al. (2016) An assessment of the potential impact of fortification of staples and condiments on micronutrient intake of young children and women of reproductive age in Bangladesh. Nutrients 8, 541.CrossRefGoogle ScholarPubMed
Madhari, RS, Boddula, S, Ravindranadh, P et al. (2020) High dietary micronutrient inadequacy in peri-urban school children from a district in South India: potential for staple food fortification and nutrient supplementation. Matern Child Nutr 16, e13065.CrossRefGoogle ScholarPubMed
Melo, L, Ng, C, Tsang, R et al. (2020) Development of novel vitamin B12 fortified yogurts using isolated and microencapsulated vitamin B12. Proc Nutr Soc 79, E264.CrossRefGoogle Scholar
Vora, RM, Alappattu, MJ, Zarkar, AD et al. (2021) Potential for elimination of folate and vitamin B12 deficiency in India using vitamin-fortified tea: a preliminary study. BMJ Nutr Prev Health 4, 293.CrossRefGoogle ScholarPubMed
Vora, RM, Antony, AC (2022) The unresolved tragedy of neural-tube defects in India: the case for folate-and vitamin-B12-fortified tea for prevention. J Indian Assoc Pediatr Surg 27, 1.CrossRefGoogle Scholar
Artés-Hernández, F, Formica-Oliveira, AC, Artés, F, Martínez-Hernández, GB (2017) Improved quality of a vitamin B12-fortified ‘ready to blend’fresh-cut mix salad with chitosan. Food Sci Technol Int 23, 513528.CrossRefGoogle ScholarPubMed
Tapola, N, Karvonen, H, Niskanen, L, Sarkkinen, E (2004) Mineral water fortified with folic acid, vitamins B6, B12, D and calcium improves folate status and decreases plasma homocysteine concentration in men and women. Eur J Clin Nutr 58, 376385.CrossRefGoogle ScholarPubMed
Zant, A, Awwad, HM, Geisel, J et al. (2019) Vitamin B12-fortified toothpaste improves vitamin status in elderly people: a randomized, double-blind, placebo-controlled study. Aging Clin Exp Res 31, 18171825.CrossRefGoogle ScholarPubMed
Siebert, A-K, Obeid, R, Weder, S et al. (2017) Vitamin B-12–fortified toothpaste improves vitamin status in vegans: a 12-wk randomized placebo-controlled study. Am J Clin Nutr 105, 618625.CrossRefGoogle ScholarPubMed
Dullemeijer, C, Souverein, OW, Doets, EL et al. (2012) Systematic review with dose-response meta-analyses between vitamin B-12 intake and European Micronutrient Recommendations Aligned’s prioritized biomarkers of vitamin B-12 including randomized controlled trials and observational studies in adults and elderly persons. Am J Clin Nutr 97, 390402.CrossRefGoogle ScholarPubMed
Buesing, S, Costa, M, Schilling, JM, Moeller-Bertram, T (2019) Vitamin B12 as a treatment for pain. Pain Physician 22, E45E52.CrossRefGoogle ScholarPubMed
Julian, T, Syeed, R, Glascow, N et al. (2020) B12 as a treatment for peripheral neuropathic pain: a systematic review. Nutrients 12, 2221.CrossRefGoogle ScholarPubMed
Banihani, SA (2017) Vitamin B12 and semen quality. Biomolecules 7, 42.CrossRefGoogle ScholarPubMed
Caballero, M, Lukawska, J, Lee, T, Dugué, P (2007) Allergy to vitamin B12: two cases of successful desensitization with cyanocobalamin. Allergy 62, 13411342.CrossRefGoogle ScholarPubMed
Tordjman, R, Genereau, T, Guinnepain, MT et al. (1998) Reintroduction of vitamin B12 in 2 patients with prior B12-induced anaphylaxis. Eur J Haematol 60, 269270.CrossRefGoogle Scholar
James, J, Warin, RP (1971) Sensitivity to cyanocobalamin and hydroxocobalamin. Br Med J 2, 262.CrossRefGoogle ScholarPubMed
Morales-Gutierrez, J, Díaz-Cortés, S, Montoya-Giraldo, MA, Zuluaga, AF (2020) Toxicity induced by multiple high doses of vitamin B12 during pernicious anemia treatment: a case report. Clin Toxicol 58, 129131.CrossRefGoogle ScholarPubMed
Veraldi, S, Benardon, S, Diani, M, Barbareschi, M (2018) Acneiform eruptions caused by vitamin B12: a report of five cases and review of the literature. J Cosmet Dermatol 17, 112115.CrossRefGoogle ScholarPubMed
Campbell, A, Heydarian, R, Ochoa, C et al. (2018) Single arm phase II study of oral vitamin B12 for the treatment of musculoskeletal symptoms associated with aromatase inhibitors in women with early stage breast cancer. Breast J 24, 260268.CrossRefGoogle ScholarPubMed
USDA (2019) FoodData Central [USDo Agriculture, editor]. https://fdc.nal.usda.gov/.Google Scholar
Probst, Y (2009) Nutrient Compostion of Chicken Meat. Australia Rural Industries Research and Development Corporation.Google Scholar
Orkusz, A (2021) Edible insects versus meat—nutritional comparison: knowledge of their composition is the key to good health. Nutrients 13, 1207.CrossRefGoogle ScholarPubMed
Roe, M, Church, S, Pinchen, H, Finglas, P (2013) Nutrient analysis of fish and fish products: analytical report. Norwich, United Kingdom: Institute of Food Research.Google Scholar
Gahruie, HH, Eskandari, MH, Mesbahi, G, Hanifpour, MA (2015) Scientific and technical aspects of yogurt fortification: a review. Food Sci Hum Wellness 4, 18.CrossRefGoogle Scholar
Roe, M, Church, S, Pinchen, H, Finglas, P (2013) Nutrient analysis of eggs: analytical report. Norwich, United Kingdom: Institute of Food Research.Google Scholar
Oosterink, JE, Naninck, EF, Korosi, A et al. (2015) Accurate measurement of the essential micronutrients methionine, homocysteine, vitamins B6, B12, B9 and their metabolites in plasma, brain and maternal milk of mice using LC/MS ion trap analysis. J Chromatogr B Analyt Technol Biomed Life Sci 998-999, 106113.CrossRefGoogle ScholarPubMed
Jiang, X, Wang, Y, Liu, J (2022) Simultaneous determination of four cobalamins in rat plasma using online solid phase extraction coupled to high performance liquid chromatography-tandem mass spectrometry: application to pentylenetetrazole-induced seizures in Sprague-Dawley rats. PLoS One 17, e0269645.CrossRefGoogle ScholarPubMed
Kahoun, D, Fojtíková, P, Vácha, F et al. (2022) Development and validation of an LC-MS/MS method for determination of B vitamins and some its derivatives in whole blood. PLoS One 17, e0271444.CrossRefGoogle ScholarPubMed
Schwertner, HA, Valtier, S, Bebarta, VS (2012) Liquid chromatographic mass spectrometric (LC/MS/MS) determination of plasma hydroxocobalamin and cyanocobalamin concentrations after hydroxocobalamin antidote treatment for cyanide poisoning. J Chromatogr B Analyt Technol Biomed Life Sci 905, 1016.CrossRefGoogle ScholarPubMed
Mandal, SM, Mandal, M, Ghosh, AK, Dey, S (2009) Rapid determination of vitamin B2 and B12 in human urine by isocratic liquid chromatography. Anal Chim Acta 640, 110113.CrossRefGoogle ScholarPubMed
Korpeti, A, Manousi, N, Kabir, A et al. (2023) Investigating the applicability of polar fabric phase sorptive extraction for the HPLC quantitation of salivary vitamin B(12) following administration of sublingual tablets and oral sprays. Talanta 258, 124482.CrossRefGoogle ScholarPubMed
Singh, R, Jaiswal, S, Singh, K et al. (2018) Biomimetic polymer-based electrochemical sensor using methyl blue-adsorbed reduced graphene oxide and functionalized multiwalled carbon nanotubes for trace sensing of cyanocobalamin. ACS Appl Nano Mater 1, 46524660.CrossRefGoogle Scholar
Wiesholler, LM, Genslein, C, Schroter, A, Hirsch, T (2018) Plasmonic enhancement of NIR to UV upconversion by a nanoengineered interface consisting of NaYF(4):Yb,Tm nanoparticles and a gold nanotriangle array for optical detection of vitamin B12 in serum. Anal Chem 90, 1424714254.CrossRefGoogle Scholar
Ahmad, M, Mohsin, M, Iqrar, S et al. (2018) Live cell imaging of vitamin B12 dynamics by genetically encoded fluorescent nanosensor. Sens Actuators B Chem 257, 866874.CrossRefGoogle Scholar
Pourreza, N, Mirzajani, R, Burromandpiroze, J (2017) Fluorescence detection of vitamin B12 in human plasma and urine samples using silver nanoparticles embedded in chitosan in micellar media. Anal Methods 9, 40524059.CrossRefGoogle Scholar
Li, D, Yuan, Q, Yang, W et al. (2018) Efficient vitamin B12-imprinted boronate affinity magnetic nanoparticles for the specific capture of vitamin B12. Anal Biochem 561-562, 1826.CrossRefGoogle ScholarPubMed
LSBio All species Vitamin B12/Cyanocobalamin Elisa Kit (Competitive EIA). https://www.lsbio.com/elisakits/manualpdf/ls-f5132.pdf.Google Scholar
abbexa Cyanocobalamin (Vitamin B12) ELISA Kit. https://www.abbexa.com/documents/manual/abx257137_ifu.pdf.Google Scholar
Epitope Diagnostics I Human Vitamin B12 CLIA Kit. https://www.epitopediagnostics.com/products/skt-070r.Google Scholar
Figure 0

Fig. 1. Structure of vitamin B12: Natural forms include 5′-deoxyadenosylcobalamin (AdoCbI), methylcobalamin and hydroxocobalamin, industrially produced is cyanocobalamin(7). Structure was created by ChemDraw, version 20.0.

Figure 1

Table 1. Cobalamin contents in selected foodstuffs

Figure 2

Fig. 2. Absorption of vitamin B12 via the IF pathway: Dietary protein-bound vitamin B12 can bind to transcobalamin I (TCI) only after its release mediated by pepsin and hydrochloric acid produced by the gastric mucosa. In the duodenum, TCI is degraded by pancreatic proteases and free cobalamin binds to intrinsic factor (IF). The IF–cobalamin complex is absorbed in the distal ileum by receptor-mediated endocytosis enabled by cubilin with participation of other protein(s), e.g. amnionless (AMN). IF is degraded in the lysosome and released cobalamin enters the cytoplasm likely by use of the transmembrane protein LMBD1. The precise mechanism of vitamin B12 efflux from enterocytes into the circulation is not yet well described. It appears to be mediated by several exporters; one of them is multidrug resistance protein 1 (MRP1, shown in teal colour).

Figure 3

Fig. 3. Physiological function of vitamin B12 and its connection with folate metabolism: (A) Together with folic acid (vitamin B9), methylcobalamin as a cofactor for the enzyme methionine synthase is necessary for the formation of methionine. During the reaction, the methyl group is transferred from methyltetrahydrofolate (CH3-THF) to homocysteine by the enzyme; the resulting tetrahydrofolate can be then converted to methylenetetrahydrofolate (CH2=THF), the form required for de novo thymidine synthesis. (B) In the conversion of methylmalonyl-coenzyme A to succinyl-coenzyme A, B12 is involved in its active form adenosylcobalamin as a cofactor of the enzyme methylmalonyl-coenzyme A mutase. The resulting succinyl-coenzyme A is a major mediator of the tricarboxylic acid (TCA) cycle; CoA, coenzyme A; DHF, dihydrofolate; THF, tetrahydrofolate.

Figure 4

Fig. 4. Formation of methylcobalamin: The highly nucleophilic cob(I)alamin reacts with a methylating agent to form methylcobalamin. Modified in ChemDraw, version 20.0 on the basis of publication of Kräutler(579).

Figure 5

Fig. 5. Formation of adenosylcobalamin: Adenosylcobalamin functions as a reversible source of the 5′-deoxyadenosyl radical, this reaction produces cob(II)alamin. Modified in ChemDraw, version 20.0 on the basis of publication of Kräutler(579).

Figure 6

Table 2. Summary of analytical methods for the assessment of vitamin B12 in biological fluids

Figure 7

Table 3. The reference intervals of the individual biomarkers, values indicating transitional status and B12 deficiency