You are on page 1of 37

TUGAS RESUME

METABOLISME DAN ZAT GIZI

Nama : Andi Kumala

Nim : J1A120003

Kelas : Gizi Kesehatan Masyarakat

Judul Jurnal Internasional :

1. Cross-Sectional Associations Between Dietary Antioxidant Vitamins


C, E and Carotenoid Intakes and Sarcopenic Indices in Women
Aged 18–79 Years
2. Metabolism of Dietary and Microbial Vitamin B Family in the Regulation of Host
Immunity

Vitamin adalah zat gizi mikro yang memiliki efek fisiologis pada berbagai
respons biologis, termasuk kekebalan inang. Oleh karena itu, kekurangan vitamin
menyebabkan peningkatan risiko mengembangkan penyakit menular, alergi, dan
inflamasi. Karena vitamin B disintesis oleh tanaman, ragi, dan bakteri, tetapi tidak
oleh mamalia, mamalia harus memperoleh vitamin B dari sumber makanan atau
mikroba, seperti mikrobiota usus. Demikian pula, beberapa bakteri usus tidak
dapat mensintesis vitamin B dan harus mendapatkannya dari makanan inang atau
dari bakteri usus lain untuk pertumbuhan dan kelangsungan hidup mereka. Hal ini
menunjukkan bahwa komposisi dan fungsi mikrobiota usus dapat mempengaruhi
penggunaan vitamin B inang dan, dengan perluasan, kekebalan inang.

Meskipun vitamin B makanan umumnya diserap melalui usus kecil,


vitamin B bakteri diproduksi dan diserap terutama melalui usus besar (10,11),
menunjukkan bahwa vitamin B yang diturunkan dari makanan dan usus mungkin
ditangani secara berbeda oleh tubuh manusia. Vitamin B adalah kofaktor dan
koenzim penting dalam beberapa jalur metabolisme, dan baru-baru ini dilaporkan
bahwa vitamin B juga memainkan peran penting dalam pemeliharaan homeostasis
imun.

Regulasi imunologi yang dimediasi vitamin B spesifik untuk sel imun dan
respons imun yang berbeda: yaitu, vitamin B yang berbeda diperlukan untuk
respons imun yang berbeda.Pernah terpikir bahwa vitamin B hanya diperoleh dari
makanan; namun, kita tahu sekarang bahwa ini bukan masalahnya dan bahwa
mikrobiota usus juga merupakan sumber vitamin yang penting. Di dalam
mikrobiota usus, tidak semua bakteri menghasilkan vitamin B dan beberapa
bakteri menggunakan vitamin B makanan atau vitamin B yang diproduksi oleh
bakteri usus lain untuk kebutuhan mereka sendiri; oleh karena itu, mungkin ada
persaingan antara inang dan mikrobiota usus untuk vitamin B
komposisi dan fungsi mikrobiota usus. Oleh karena itu,imunologi yang
diperantarai vitamin pemeliharaan juga bervariasi antar individu. Lebih jauh
pemeriksaan di bidang ini diperlukan, dan informasi baru yang ditemukan akan
membantu mengembangkan era baru kesehatan dan nutrisi yang presisi.

vitamin antioksidan diet (A, C, E dan karotenoid) adalah kandidat yang


menjanjikan untuk pencegahan dan pengobatan hilangnya massa dan fungsi
terkait usia. Vitamin ini mempengaruhi otot rangka dan berfungsi melalui
perannya sebagai antioksidan eksogen dan agen anti-inflamasi. Selain itu, vitamin
C terlibat dalam sintesis kolagen dan karnitin dan retinol dalam metabolisme
protein, pembentukan kolagen, dan oksidasi lipid.

vitamin C digunakan untuk sifat antioksidan dan fungsi sistem kekebalan


tubuh. Oksalat adalah produk akhir dari metabolisme vitamin C. Asam askorbat
yang bersirkulasi diubah menjadi antioksidan jaringan, menghasilkan pembentuk
an radikal askorbat. Radikal tersebut kemudian akan diubah menjadi
dehyroascorbate, yang dapat diubah menjadi asam diketogulonat, molekul tidak
stabil yang dapat dengan mudah terurai menjadi oksalat. Peningkatan konsumsi
asam askorbat menyebabkan pembentukan oksalat yang berlebihan. Kenaikan
kecil oksalat urin dapat meningkatkan risiko pembentukan kristal kalsium oksalat,
dan kadar oksalat urin yang lebih tinggi merupakan faktor risiko utama
pembentukan batu ginjal kalsium oksala. Sayuran penyumbang vitamin C terbesar
adalah paprika, kubis Brussel dan brokoli. Untuk karoten sumber nabati utama
adalah wortel dan bayam sedangkan Buah secara keseluruhan adalah penyumbang
terbesar asupan vitamin C.

Ada relevansi mekanistik yang jelas dari vitamin antioksidan pada


kesehatan otot rangka selama penuaan yang juga mendukung temuan pengamatan
kami. Suplementasi likopen,karoten atau vitamin antioksidan campuran pada
hewan percobaan memiliki efek positif pada kekuatan otot atau aktivitas fisik atau
mengurangi stres oksidatif atau hilangnya otot rangka.vitamin C memiliki efek
perlindungan pada biomarker oksidatif, sitokin inflamasi dan CRP, vitamin C juga
merupakan bagian integral dari sintesis kolagen dan karnitin yang penting untuk
metabolisme asam lemak rantai panjang selama aktivitas fisik.

suprafisiologis vitamin C dapat digunakan untuk memperbaiki asupan


makanan rendah pada kelompok rentan yang lebih tua, ini perlu digunakan dengan
hati-hati karena efek prooksidan yang merugikan dari vitamin C dosis tinggi
jangka panjang (> 500 mg per hari) dibandingkan konsentrasi yang ditemukan
secara alami dalam makanan.
Calcified Tissue International (2020) 106:331–342
https://doi.org/10.1007/s00223-019-00641-x

ORIGINAL RESEARCH

Cross-Sectional Associations Between Dietary Antioxidant


Vitamins C, E and Carotenoid Intakes and Sarcopenic Indices in
Women Aged 18–79 Years
A. A. Welch1 · A. Jennings1 · E. Kelaiditi1,2 · J. Skinner1 · C. J. Steves3

Received: 1 August 2019 / Accepted: 22 November 2019 / Published online: 7 December 2019
© The Author(s) 2019

Abstract
The prevalence of sarcopenia is increasing in aging populations, so prevention is critical. Vitamins (A, C, E and
carotenoids) modify skeletal muscle via protein and collagen synthesis and anti-inflammatory activities. Previous studies
have not inves- tigated intake of these vitamins in relation to sarcopenic indices in both younger and older-aged women.
Indices of skeletal muscle mass (as fat-free mass (FFM) relative to body size) were measured using DXA and leg
explosive power (LEP) using the Nottingham Power Rig in 2570 women aged 18–79 years. Adjusted measures of skeletal
muscle were calculated according to quintiles of vitamin C, E, retinol and carotenoid intake, derived from Food Frequency
Questionnaires, after stratification by age. Higher vitamin C intake was associated with significantly higher indices of
FFM and LEP, (Q5-Q1 = 2.0–12.8%, P < 0.01–0.02). Intakes of total and individual carotenoids were significantly
associated with indices of FFM and LEP (Q5-Q1 = 1.0–7.5%). Vitamin E was significantly associated with FFM% and
FFMBMI only. In mutually adjusted analysis with vitamin C, total carotene, vitamin E and protein in the model, the
strongest associations were with vitamin C. These associations were stronger in younger women (< 65 years). For the first
time, our research shows higher dietary intakes of antioxidant vitamins, particularly vitamin C, is associated with higher
skeletal muscle mass and power in free-living women. These findings have relevance for the treatment and prevention of
frailty and sarcopenia throughout adulthood.

Keywords Sarcopenia · Diet · Vitamin C · Vitamin E · Carotenes · Vitamin A · Skeletal muscle · Grip strength

Background and Aims


and mortality, leading to longer and more expensive hospi-
Across the world, populations are aging rapidly leading to tal stays. These conditions also impact adversely on qual-
increased prevalence of sarcopenia and frailty, conditions ity of life for individuals [1]. The estimated prevalence of
that also increase the risk of osteoporosis, falls, fractures sarcopenia in the community is 1–29% in people over the
age of 60 years, which rises to 17.7–87% in those living in
assisted or residential or care facilities [1–3]. Frailty is esti-
mated to affect 25% of people over the age of 80 years. We
Electronic supplementary material The online version of this need, therefore, strategies to prevent the gradual age-
article (https://doi.org/10.1007/s00223-019-00641-x) contains
related decline in skeletal muscle mass and function, or
supplementary material, which is available to authorized
users. sarcopenic indices, that start in middle age to maintain
intrinsic capacity in older adults [4].
 A. A. Welch The established mechanisms of aging of skeletal muscle
a.welch@uea.ac.uk
include increases in circulating cytokines and production
1
Department of Epidemiology & Public Health, Norwich of reactive oxygen species (ROS), with age which have
Medical School, University of East Anglia, Norwich detri- mental effects on synthesis of protein as well as direct
Research Park, Norwich, Norfolk NR4 7TJ, UK cellular damage of skeletal muscle fibres and DNA [5].
2
Faculty of Sport, Health and Applied Science, St Additionally, there are changes to the proportion, quality
Mary’s University, Waldegrave Road, and viscoelastic properties of the different types of
Twickenham, London TW1 4SX, UK collagen that form the important structural component of
3
Department of Twin Research and Genetic skeletal muscle cells, con- nective tissues and tendons
Epidemiology, King’s College, London, UK
13
during aging [6]. As endogenous

13
33 A. A. Welch et al.

antioxidant efficiency is reduced with aging, and skeletal aged 18–103 years who are representative of singleton
muscle generates the greatest quantities of ROS in the
body, exogenous antioxidant vitamins have potential
importance for skeletal muscle health.
Dietary antioxidant vitamins (A, C, E and carotenoids)
are, therefore, promising candidates for the prevention and
treat- ment of age-related loss of mass and function. These
vitamins influence skeletal muscle and function through
their roles as exogenous antioxidant and anti-inflammatory
agents. In addi- tion, vitamin C is involved in collagen and
carnitine synthesis and retinol in protein metabolism,
collagen formation, and lipid oxidation [7, 8].
To date, dietary treatments for sarcopenia and frailty
have focused largely on interventions with protein intake,
with or without resistance exercise, which is important for
skeletal muscle but the effectiveness of intervention studies
in indi- viduals with sarcopenia and frailty have been
mixed [9, 10]. As there are no effective pharmacological
treatments for sar- copenia, frailty and the loss of skeletal
muscle mass and func- tion with age, identifying other
dietary factors to prevent or attenuate losses of muscle
mass and function in middle and early old age is important.
Whilst limited previous research has studied nutritional
intake or blood concentrations of vitamin A, C, E or carot-
enoids and measures of skeletal muscle mass or function in
older-aged populations, none has measured their relative
effec- tiveness in relation to a range of indices of both
skeletal mass and function in young as well as in older-aged
women [11–19]. Therefore, the purpose of this study was to
first, understand the associations between: (i) dietary
vitamins C, E and A and (ii) the full range of dietary
carotenoids; α-carotene, β-carotene, β-cryptoxanthin,
lycopene, lutein and zeaxanthin and indices of skeletal
muscle mass [i.e. Fat Free Mass Index (FFMI), percentage
fat-free mass (FFM%) and fat-free mass adjusted for body
mass index (FFMBMI)], and function [i.e. hand grip
strength, arm muscle quality and leg explosive power
(LEP)] in a population of women with a wide age range.
Second, to understand whether these associations differed
in young women compared with those over the age of 65
years, third to estimate the relationship between the
inflammatory cytokine C-reactive protein (CRP) and indices
of skeletal muscle health and fourthly, to determine the
relative associations of vitamins A, C, E, total carotene and
protein by including all the nutri- ents in the same
statistical models to determine which of the vitamins was
the most strongly associated with the indices of skeletal
muscle health.

Methods

The TwinsUK registry is an ongoing population study


of approximately 12,000 male and female (83%) twins,

13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 33
Intakes…
populations in the United Kingdom [20]. Data were used
from 2570 women who had completed a food frequency
questionnaire (FFQ) and attended for dual-energy X-ray
absorptiometry (DXA) measurements between 1996 and
2000. Within this group, there were 1914 individuals with
measures of leg explosive power and 1658 individuals
with measures of high sensitivity C-reactive protein (hs-
CRP). Between 2005 and 2008, 949 women completed an
FFQ and had grip strength measured (including 512
individu- als from the first cohort) (Supplementary Fig.
1). Ethical approval was obtained from St. Thomas’s
Hospital Research Ethics Committee and informed
consent was acquired from all participants.

Dietary Intake

Participants completed a 131 item-validated FFQ with


nutrient values calculated using the UK national nutrient
database and a database of the carotenoid composition of
foods [21, 22]. Individuals were excluded from the
dietary analyses if answers to > 10 food items were left
blank or the ratio of estimated total energy intake to the
estimated basal metabolic rate fell 2 SDs outside the
mean ratio. Energy- reporting quality was estimated using
the ratio of reported energy intake (EI) to estimated
energy expenditure (EER), the EI:EER ratio expressed as
the percentage of EI to EER, was calculated and was
included as a covariate for adjust- ment in the statistical
analyses [23].

Muscle Mass, Strength and Power

Fat-free mass (FFM) was measured by DXA scans (Hol-


ogic QDR-2000 DXA scanner, Hologic Inc., Waltham,
MA, USA) and adjusted to body size using the following
cal- culations: FFMI: FFM (kg)/ height (m2) [1], FFM%:
FFM (kg)/ weight (kg) *100 and FFMBMI: FFM (kg)/ BMI
(kg/ m2). FFM (kg) divided by BMI (kg/m2) (FFMBMI)
takes into account the increase in body size, scaled for
height and adjusting FFM for BMI is likely to set
meaningful cut- off points for clinical use [1]. Isometric
grip strength was assessed using a Jamar hand grip
dynamometer (Sammons, Preston, UK) on the dominant
arm with reproducibility assessed by repeated
measurement on 24 individuals (CV of 11.4%) [24] and
adjusted for mean arm lean mass as arm muscle quality
(grip strength (kg)/ mean arm lean mass (kg). LEP was
measured using the Nottingham Power Rig which
assesses the force and velocity of muscle contraction
from the quadriceps, normalised for body weight (LEP
(watts)/ weight (kg)) [25]. LEP is validated with high
reliability (reli- ability coefficient 0.97, coefficient of
variation 9.4%, over one week period in adults) [25].
13
33 A. A. Welch et al.

C-reactive Protein smoking status (never, former, current), energy

Circulating hs-CRP was measured by a highly sensitive


automated microparticle capture enzyme immunoassay,
standardised on the World Health Organisation
International Reference Standard for CRP immunoassay as
previously described [26].

Covariates

Information on lifestyle, medication use, menopausal


status, and demographic variables were obtained using
standardised nurse-administered questionnaires. Weight
and height were measured to the nearest 0.1 kg and to the
nearest 0.5 cm, respectively. BMI was calculated as weight
in kilograms (kg) divided by height in meters squared (m2).
Leisure- and work-time physical activity was self-reported
using a ques- tionnaire significantly correlated with in
depth assessments of physical activity in a subset of this
cohort [27]. For par- ticipants in the grip strength group (n
= 949 women) who had missing data on physical activity
(N = 10), the General Practice Physical Activity
Questionnaire (GPPAQ) was used. This was based on the
original physical activity index developed by the European
Prospective Investigation into Cancer and Nutrition (EPIC)
cohort. The EPIC physical activity index was previously
validated against heart rate monitoring in two independent
studies [28].

Statistical Analyses

Descriptive statistics (means ± SDs or % (n)) were


analysed for all participants and for those under 65 years (n
= 2346) and ≥ 65 years (n = 224). Multivariate regression
analysis was used to calculate statistical trends and
adjusted val- ues (least square means) for FFM%, FFMI,
FFMBMI, LEP, arm muscle quality and hs-CRP (outcomes)
classified by quintile of dietary vitamins C, E and A and
dietary carot- enoids; α-carotene, β-carotene, β-
cryptoxanthin, lycopene, lutein and zeaxanthin (exposures).
The percentage difference between quintiles 1 and 5 was
calculated as a percentage of the value in quintile 1. As
physical activity and smoking habit were related to indices
of skeletal muscle the analy- ses were adjusted for physical
activity and smoking habit. All models were adjusted for
age (years), physical activity (active, moderately active,
inactive), smoking status (never, former, current), energy
intake (kcal/d) and potential mis- reporting of energy
intake (EI:EER). FFMI was adjusted for age (years),
physical activity (active, moderately active, inactive),

13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 33
Intakes… 2
intake (kcal/d), potential mis-reporting of energy intake trend < 0.001), FFMI was 0.4 kg/ m higher (± 0.1 P-trend
(EI:EER) and fat mass (kg). LEP and arm muscle = 0.002), FFMBMI was 0.03 kg/kg/ m2 higher (± 0.01 P-
qual- ity were adjusted for age (years), physical activity trend = 0.023) and LEP was 10.9 w/ kg higher (± 2.6 P-
(active, moderately active, inactive), smoking status trend < 0.001), when comparing extreme
(never, former, current), energy intake (kcal/d), potential
mis-reporting of energy intake (EI:EER) and menopausal
status (pre-meno- pausal/ post-menopausal), use of HRT
(yes/no) and height (m). High sensitivity (hs) CRP was
adjusted for age (years), physical activity (active,
moderately active, inactive), smok- ing status (never,
former, current), energy intake (kcal/d), potential mis-
reporting of energy intake (EI:EER), and BMI, use of
anti-inflammatory medications (yes/no) and HRT
(yes/no). Values for hs-CRP were skewed and therefore,
natural log-transformed values were used for the
analyses.
In analysis stratified by age, we also assessed the
associa- tions of FFM%, FFMI, FFMBMI and LEP with
quintiles of intake for those under 65 years (n = 2346)
and tertiles for those ≥ 65 years (n = 224). The data for
those ≥ 65 years was analysed for tertiles due to the
smaller number of women in this group. For these and
the comparative analyses we only considered vitamins A,
C, E, total carotene and protein intakes as these
demonstrated the greatest associations, as shown in Table
2. In additional analyses, to compare the relative
associations of vitamins A, C, E, total carotene and
protein we included all these nutrients in the same statisti-
cal models and standardised the values for FFM%, FFMI,
FFMBMI and LEP using Z-scores. Values for the FFM
indi- ces and LEP were also adjusted for covariates as
described previously, and as in the models shown in
Table 3.
Values in the text are means ± SE. A P value < 0.05
was considered statistically significant. P trend was
calculated from the multivariable regression across
quintiles of intake of the nutrients. All analyses were
performed with Stata statistical software version 14.0
(Stata Corp, College Sta- tion, TX) and included the
robust cluster regression option in STATA to account for
clustering within twin pairs.

Results

The characteristics of the participants are presented in


Table 1, stratified by age (< 65 years and ≥ 65 years). The
mean age (±SD) of participants was 48.37 ± 12.7 years
and mean (± SD) BMI was 24.9 ± 4.14 kg/m 2. More than
one- half of the participants were moderately active
(53.9%), and 18.2% were current smokers. In
multivariable analyses, women in the highest quintile of
vitamin C intake had sign- ficantly higher FFM%, FFMI,
FFMBMI and LEP, compared to those in the lowest quintile
(Table 2). Specifically, FFM% was 1.4% higher (± 0.4 P-

13
33 A. A. Welch et al.

intake quintiles. In terms of vitamin E, FFM% (Q5-Q1


1.6% ± 0.5 P-trend = 0.002) and FFMBMI (Q5-Q1 0.06 ± Age-Stratified Analyses
0.02
P-trend = 0.002) were positively associated with intakes. In age-stratified analyses, we observed that associations
For total carotene, associations were also significant for between vitamin C and E with FFM% were observed in
FFM% (Q5-Q1 0.6% ± 0.4 P-trend = 0.028), FFMI (Q5- the younger (< 65 years) but not the older (≥ 65 years)
Q1 0.2 ± 0.1 subset (Table 3). Specifically, vitamin C was associ-
P-trend = 0.012) and LEP (Q5-Q1 6.4 ± 2.7 P-trend = ated with a 0.3% (95% CI 0.1, 0.5 P-trend = 0.002) and
0.011). vitamin E with a 0.4% (95% CI 0.2, 0.7 P-trend = 0.001)
The results of the associations between the nutrients and higher FFM% per quintile of intake. Likewise, associa-
arm muscle quality were non-significant ranging from a tions between vitamin C (2.3 95% CI 1.1, 3.6 per quintile
beta coefficient (95% CI) of − 0.075 (− 0.199, 0.048) per P-trend < 0.001) and total carotene (1.7 95% CI 0.4, 2.9
quintile for vitamin C intake (P-trend = 0.231) to 0.036 (− per quintile P-trend = 0.009) and LEP (w/kg) were only
0.150, 0.222) per quintile for vitamin E intake (P-trend = observed in participants aged < 65 years. Similar trends
0.705). The results of the associations between the were also found with FFMI and FFM/BMI, although there
nutrients and hs-CRP were also non-significant ranging was no significant association between FFM/BMI and vita-
from a beta coef- ficient (95% CI) of − 0.008 (− 0.046, min C in those < 65 years.
0.029) per quintile for vitamin C intake (P-trend = 0.666)
to − 0.023 (− 0.070, 0.025) per quintile for vitamin E
intake (P-trend = 0.349). Also no associations were
observed for vitamin A (measured as retinol). Standardised Analyses

In standardised analyses, we compared the relative


Individual Carotenoids associations of vitamins C, E, retinol and total caro-
tene with FFM% and LEP (Figs. 1, 2). Associations
Intakes of all the individual carotenoids, with the excep- between vitamin C and FFM% and LEP were maintained
tion of leutein + zeaxanthin, were significantly associated after mutual adjustment for the other nutrients, includ-
with FFMI. The strongest association was observed for α- ing protein (FFM% 0.17 SD ± 0.07 P-trend = 0.013
carotene intake (Q5-Q1 0.24 kg/m 2 ± 0.1 P-trend = 0.03, and LEP 0.27 SD ± 0.08 P-trend = 0.004). A signifi-
1.6%). Significant associations were also found with FFM cant negative association with protein was observed for
% for ß-cryptoxthanin and with FFM% and FFMBMI for FFM% (− 0.18 SD ± 0.06 P-trend < 0.001), FFMBMI
lutein and zeaxthanin, with interquintle differences – 0.27 ± 0.06 P-trend < 0.001 and LEP (− 0.14 SD ± 0.07
ranging from P-trend = 0.007) but the association with FFMI was not
1.1 to 7.2%. LEP was associated significantly with the
carot- enoids, with the exception of α-carotene, with
differences ranging from 6.3 to 7.5% when comparing
extreme quintiles of intake.

Fig. 1 The relative associa- P-


tions of vitamin C, vitamin E, tren 0.7
retinol, carotene and protein d< 0.6
with percentage fat-free mass 0.05 0.5
in 2570 females aged 18–79
0.4
years, stratified by age1. Values
repre- sent the difference in 0.3
stand- 0.2
ardised values of percentage 0.1
fat-free mass between
SD

partici- 0
pants in Q5 vs Q1 of intake -0.1
(T3- T1 for > 65 years sub- -0.2
group) with all nutrients
-0.3
included in the model. Values
were also adjusted for age, -0.4
physical activity, smoking -0.5
status, energy intake and
-0.6
underreporting. *

13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 33
Intakes…
-0.7 Protein

* *

* *

A
ll
(n
=
2
5
7
0)

<
6
5
y
(n
=
2
3
4
6)


6
5
y
(n
=
2
2
4)
Vita
min
C

Vita
min
E

Tot
al
caro
tene

Reti
nol

13
33 A. A. Welch et al.

Table 1 Characteristics and


dietary intakes of n = 2570
C haracteristic All < 65 years ≥ 65 years
females aged 18–79 years,
stratified by agea n= 2570 2346 224
Age (years) 48.3 ± 12.7 46.5 ± 11.6 68.0 ± 2.81
BMI (kg/m2) 24.9 ± 4.14 24.8 ± 4.11 26.1 ± 4.23
Fat mass (kg) 22.7 ± 7.87 22.5 ± 7.90 25.0 ± 7.26
Fat free mass (%) 61.1 ± 6.50 61.4 ± 6.46 58.0 ± 6.08
Fat Free Mass Index (kg/m2) 15.0 ± 1.72 15.0 ± 1.71 15.0 ± 1.80
Fat-free mass/ BMI (kg/ kg/m2) 1.62 ± 0.23 1.63 ± 0.23 1.47 ± 0.19
Leg explosive powerb (watts) 89.8 ± 36.8 91.3 ± 36.7 67.5 ± 30.8
Leg explosive powerb (watts/kg) 90.9 ± 36.5 92.4 ± 36.3 68.3 ± 31.6
hs-CRPc (mg/L) 2.49 ± 2.30 2.47 ± 2.30 2.72 ± 2.26
Grip strengthd (kg) 28.8 ± 5.95 30.1 ± 5.66 25.3 ± 5.20
Energy intake (kcal/d) 1979 ± 524 1978 ± 533 1989 ± 415
Protein (% energy) 16.6 ± 2.62 16.6 ± 2.62 16.9 ± 2.68
Vitamin C (mg/d) 155 ± 80.2 154 ± 80.4 165 ± 78.3
Vitamin E (mg/d) 11.4 ± 4.57 11.3 ± 4.60 11.7 ± 4.26
Total carotene (µg/d) 3448 ± 1944 3416 ± 1964 3777 ±
1688
α-Carotene (µg/d) 559 ± 416 554 ± 418 621 ± 389
β-Carotene (µg/d) 3091 ± 1757 3062 ± 1775 3398 ±
1529
Retinol (µg/d) 559 ± 789 545 ± 793 700 ± 728
β-Cryptoxanthin (µg/d) 200 ± 194 201 ± 197 192 ± 166
Lycopene (µg/d) 1347 ± 958 1364 ± 968 1165 ± 820
Lutein + zeaxanthin (µg/d) 2267 ± 1478 2253 ± 1501 2413 ±
1204
Underreporting (EI:EER, %) 87.4 ± 24.6 86.7 ± 24.6 95.0 ± 24.2
Physical activity (active, %) 24.2% (622) 23.9% (561) 27.2% (61)
Moderately active (%) 53.9% (1385) 54.2% (1271) 50.9% (114)
Inactive (%) 21.9% (563) 21.9% (514) 21.9% (49)
Smoking status (current, %) 18.2% (468) 19.1% (449) 8.48% (19)
Menopausal status (post-menopausal, %) 47.4% (1218) 42.5% (997) 98.7% (221)
Anti-inflammatory medicationc (yes, %) 6.15% (102) 6.06% (90) 6.98% (12)
Hormone replacement therapyc (yes, %) 6.33% (105) 6.33% (94) 6.40% (11)

EI:EER ratio of reported energy intake to estimated energy requirements


a
Values are mean ± SD or % (n), n = 2570
Values for a subset of b1914
c
1658
d
949 participants

significant 0.05 SD ± 0.06 P-trend = 0.390. The results vitamin E, the food groups cakes and biscuits and whole
for vitamin E, not shown in the figures were: FFMI grain cereals contributed more to intake than vegetables.
– 0.05 SD ± 0.08 P-trend = 0.487 and FFMBMI 0.19 ± 0.09
P-trend = 0.025.
Total vegetable intake was a significant source of vita-
mins C, E and carotene intake, however, the specific veg-
etables that contributed differed for each nutrient (Fig. 3).
For vitamin C the main contributors were peppers, Brus-
sels sprouts and broccoli, for vitamin E avocado, mush-
rooms and spinach, and for carotene, carrots and spinach.
Fruit intake also contributed to vitamin C intake, and for

13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 33
Intakes…
Discussion

Our findings of significant associations between higher


intakes of vitamin C, total carotene and specific carote-
noids and improved sarcopenic indices of skeletal
muscle; FFM and LEP were found in a cohort of women
with a wide age range (18–79 years). The scale of the
associa- tions for these vitamins ranged from 1.0 to 3.2%
for indi- ces of FFM and from 6.3 to 12.8% for LEP,
comparing the highest and lowest intakes and adjusting
for relevant covariates including age. This highlights the
potential clin- ical importance of the findings as the
yearly losses of FFM

13
33 A. A. Welch et al.

Table 2 Fat free mass indices


Intakeb FFM (%) FFM/BMI FFMI (kg/mb) LEPc (w/kg)
and leg explosive power by
quintile of nutrient intake in Vitamin C (mg/d)
2570 females aged 18–79
yearsa Q1 68.8 ± 16.4 60.4 (59.8,60.9) 1.60 (1.58,1.62) 14.8 (14.7,15.0) 85.2 (81.8,88.6)
Q2 108 ± 9.43 60.9 (60.4,61.4) 1.61 (1.59,1.63) 15.0 (14.9,15.2) 90.8 (87.2,94.4)
Q3 141 ± 10.1 61.1 (60.6,61.6) 1.62 (1.60,1.64) 15.0 (14.9,15.1) 90.1 (86.4,93.8)
Q4 181 ± 13.8 61.3 (60.8,61.8) 1.62 (1.60,1.64) 15.1 (14.9,15.2) 92.0 (88.2,95.9)
Q5 276 ± 80.3 61.8 (61.3,62.3) 1.63 (1.61,1.65) 15.2 (15.1,15.3) 96.1 (92.3,99.9)
P-trend – < 0.01 0.02 < 0.01 < 0.01
Q5-Q1% – 2.36 1.99 2.40 12.79
Vitamin E (mg/d)
Q1 5.90 ± 1.16 60.3 (59.7,60.9) 1.58 (1.56,1.61) 14.9 (14.8,15.1) 89.2 (84.8,93.6)
Q2 8.59 ± 0.63 60.7 (60.2,61.2) 1.60 (1.58,1.62) 15.1 (14.9,15.2) 90.6 (86.9,94.3)
Q3 10.7 ± 0.64 61.3 (60.8,61.8) 1.63 (1.61,1.65) 15.1 (14.9,15.2) 91.9 (88.1,95.8)
Q4 13.3 ± 0.81 61.2 (60.7,61.8) 1.62 (1.60,1.64) 15.0 (14.9,15.2) 90.9 (87.3,94.5)
Q5 18.3 ± 3.39 61.9 (61.2,62.5) 1.64 (1.62,1.66) 15.0 (14.8,15.2) 91.6 (87.1,96.2)
P-trend – < 0.01 < 0.01 0.70 0.54
Q5-Q1% – 2.60 3.54 0.42 2.77
Total carotene (µg/d)
Q1 1342 ± 377 60.8 (60.3,61.3) 1.61 (1.59,1.63) 14.9 (14.7,15.0) 85.6 (81.8,89.3)
Q2 2379 ± 320 60.9 (60.4,61.4) 1.61 (1.59,1.63) 15.0 (14.9,15.1) 91.2 (87.7,94.8)
Q3 3223 ± 189 60.8 (60.3,61.3) 1.61 (1.59,1.63) 15.0 (14.9,15.2) 90.8 (87.0,94.5)
Q4 3991 ± 321 61.6 (61.1,62.1) 1.63 (1.61,1.65) 15.1 (15.0,15.3) 94.8 (91.1,98.4)
Q5 6303 ± 2107 61.4 (60.9,62.0) 1.62 (1.60,1.64) 15.1 (14.9,15.2) 91.9 (88.2,95.7)
P-trend – 0.03 0.11 0.01 0.01
Q5-Q1% – 1.03 0.96 1.44 7.45
α-Carotene (µg/d)
Q1 131 ± 62.8 61.1 (60.6,61.7) 1.62 (1.60,1.64) 14.9 (14.8,15.0) 88.1 (84.6,91.7)
Q2 265 ± 88.2 61.1 (60.6,61.6) 1.62 (1.60,1.64) 15.0 (14.9,15.1) 90.8 (86.9,94.8)
Q3 605 ± 9.02 60.8 (60.3,61.2) 1.60 (1.58,1.62) 15.1 (14.9,15.2) 91.8 (88.3,95.3)
Q4 638 ± 11.7 61.0 (60.5,61.6) 1.62 (1.59,1.64) 15.0 (14.9,15.2) 92.8 (89.2,96.5)
Q5 1158 ± 467 61.4 (60.9,62.0) 1.62 (1.60,1.64) 15.1 (15.0,15.3) 90.7 (86.9,94.4)
P-trend – 0.62 0.94 0.03 0.24
Q5-Q1% – − 0.46 – 0.02 – 1.61 2.85
β-Carotene (µg/d)
Q1 1194 ± 344 60.7 (60.2,61.2) 1.60 (1.58,1.63) 14.9 (14.7,15.0) 85.6 (81.8,89.4)
Q2 2139 ± 272 61.0 (60.6,61.5) 1.61 (1.60,1.63) 15.0 (14.9,15.2) 91.4 (87.8,94.9)
Q3 2861 ± 175 60.9 (60.4,61.4) 1.61 (1.59,1.63) 15.0 (14.9,15.1) 90.1 (86.4,93.8)
Q4 3589 ± 294 61.4 (60.9,61.9) 1.62 (1.60,1.64) 15.1 (15.0,15.3) 95.3 (91.6,98.9)
Q5 5673 ± 1911 61.4 (60.9,62.0) 1.62 (1.60,1.65) 15.1 (14.9,15.2) 91.9 (88.2,95.7)
P-trend – 0.05 0.17 0.01 < 0.01
Q5-Q1% – 1.20 1.23 1.50 7.38
Retinol (µg/d)
Q1 137 ± 43.1 61.2 (60.7,61.8) 1.61 (1.59,1.63) 15.1 (14.9,15.3) 92.1 (88.1,96.2)
Q2 245 ± 25.1 60.8 (60.3,61.3) 1.60 (1.58,1.62) 15.0 (14.9,15.2) 90.7 (87.0,94.5)
Q3 347 ± 36.6 61.0 (60.5,61.5) 1.61 (1.59,1.63) 15.0 (14.8,15.1) 89.7 (86.2,93.2)
Q4 639 ± 187 61.4 (60.8,61.9) 1.63 (1.60,1.65) 15.1 (15.0,15.3) 90.8 (87.2,94.3)
Q5 1426 ± 1412 61.0 (60.5,61.6) 1.62 (1.61,1.64) 14.9 (14.8,15.1) 91.0 (87.0,95.0)
P-trend – 0.85 0.14 0.23 0.78
Q5-Q1% − 0.36 0.91 – 1.30 – 1.24
β-Cryptoxanthin (µg/d)
Q1 38.3 ± 16.8 60.7 (60.2,61.2) 1.60 (1.58,1.62) 14.9 (14.8,15.0) 87.7 (84.0,91.4)
Q2 82.9 ± 12.1 61.0 (60.5,61.5) 1.62 (1.60,1.64) 15.0 (14.8,15.1) 89.1 (85.6,92.5)

13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 33
Intakes…
Table 2 (continued)
Intakeb FFM (%) FFM/BMI FFMI (kg/mb) LEPc (w/kg)
Q3 146 ± 25.2 61.0 (60.5,61.4) 1.61 (1.59,1.63) 15.1 (15.0,15.2) 90.6 (87.1,94.1)
Q4 234 ± 36.4 61.5 (61.0,62.0) 1.63 (1.61,1.65) 15.0 (14.9,15.2) 93.7 (90.2,97.3)
Q5 499 ± 230 61.4 (60.8,61.9) 1.62 (1.60,1.64) 15.1 (15.0,15.3) 93.2 (89.4,97.0)
P-trend – 0.03 0.22 0.03 < 0.01
Q5-Q1% – 1.09 0.87 1.50 6.30
Lycopene (µg/d)
Q1 412 ± 168 61.0 (60.4,61.5) 1.61 (1.59,1.63) 15.0 (14.8,15.1) 87.2 (83.7,90.7)
Q2 821 ± 99.2 61.3 (60.8,61.8) 1.63 (1.61,1.65) 15.0 (14.8,15.1) 91.5 (87.4,95.5)
Q3 1161 ± 100 61.4 (60.9,61.8) 1.63 (1.61,1.65) 15.0 (14.9,15.1) 93.3 (89.6,97.1)
Q4 1573 ± 154 61.2 (60.7,61.6) 1.62 (1.60,1.63) 15.1 (14.9,15.2) 89.8 (86.2,93.4)
Q5 2771 ± 1121 60.6 (60.1,61.2) 1.59 (1.57,1.61) 15.1 (15.0,15.3) 92.5 (88.5,96.5)
P-trend – 0.33 0.05 0.03 0.18
Q5-Q1% – – 0.58 – 1.57 1.29 6.05
Lutein + zeaxanthin (µg/d)
Q1 835 ± 261 60.5 (60.0,61.1) 1.59 (1.57,1.61) 15.0 (14.8,15.1) 88.4 (84.6,92.2)
Q2 1462 ± 153 60.6 (60.2,61.1) 1.60 (1.58,1.62) 15.0 (14.9,15.2) 87.7 (84.4,91.0)
Q3 1970 ± 152 61.1 (60.6,61.6) 1.62 (1.60,1.64) 15.0 (14.9,15.2) 90.3 (86.8,93.8)
Q4 2602 ± 216 61.3 (60.8,61.9) 1.62 (1.61,1.64) 15.1 (14.9,15.2) 93.1 (89.4,96.8)
Q5 4464 ± 1744 61.9 (61.3,62.4) 1.64 (1.62,1.66) 15.0 (14.9,15.2) 94.8 (91.0,98.6)
P-trend – < 0.01 < 0.01 0.37 < 0.01
Q5-Q1% – 2.27 3.15 0.52 7.23

FFM fat-free mass, FFMI Fat Free Mass Index and LEP leg explosive power
a
Values are adjusted means (least square means) ± SE, n = 2570. Means were adjusted for age, physical
activity, smoking status, energy intake, protein intake and underreporting and Fat Free Mass Index was
additionally adjusted for fat mass. Participant numbers by quintile were Q1 = 514; Q2 = 514; Q3 =
514;
Q4 = 514; Q5 = 514
b
Intake values are unadjusted means ± SD
c
Values are mean (least square means) ± SE, n = 1914. Means were adjusted for age, physical activ-
ity, smoking status, energy intake, protein intake, underreporting, menopausal status, hormone replace-
ment therapy and height. Participant numbers by quintile were Q1 = 383; Q2 = 383; Q3 = 383; Q4 = 383;
Q5 = 382

and strength range from 1%, for skeletal muscle mass, and
3% for grip strength, for people over the age of 50 years Different foods contributed to intake of the nutrients
[29]. Moreover, using standardised analyses to compare inves- tigated. Although vegetables were the most significant
the different vitamins and protein, the associations were source, the specific vegetables contributing to these nutrients
greatest for vitamin C, despite mutual adjustment for all differed. The largest vegetable contributors to vitamin C
the nutrients investigated in our study, including protein were peppers, Brussels sprouts and broccoli. For carotene
intake. This research highlights the importance of dietary the major veg- etable sources were carrots and spinach
associations with muscle mass in individuals of all ages, whereas vitamin E was supplied by avocado and
supporting previous research that has shown that notable mushrooms. Overall fruit was the greatest contributor to
changes in skeletal muscle mass occur earlier in adult life vitamin C intakes, and whole grain cereals were the greatest
(between 30 and 45 years of age) [30, 31]. contributor to vitamin E intake. Our findings highlight the
The range of nutrients across quintiles of intake investi- importance of eating a broad range of vegetables, fruits and
gated in our study varied from 3.1 fold for vitamin E, and whole grain cereal foods to achieve opti- mal intakes of
fourfold for vitamin C, to 4.6 fold for total carotene and vitamin C, carotenes and vitamin E.
to larger differences for retinol (10.4 fold). However, the
associations with vitamin E intake were only found with
FFM expressed either as a percentage or divided by BMI,
and no associations were found with retinol.

13
33 A. A. Welch et al.

Table 3 Relative associations between indices of fat-free mass and leg explosive power calculated according to quantile of nutrient intake in
2570 females aged 18–79 years, stratified by agea

All (n = 2570) < 65 years (n = 2346) ≥ 65 years (n = 224)


Quintile of nutrient intake β (95% CI) P-trend β (95% CI) P-trend β (95% CI) P-trend

FFM (%)
Vitamin C (mg/d) 0.28 (0.1, 0.5) < 0.01 0.30 (0.1, 0.5) < 0.01 0.50 (− 0.5, 1.5) 0.32
Vitamin E (mg/d) 0.41 (0.2, 0.6) < 0.01 0.41 (0.2, 0.7) < 0.01 0.31 (− 0.7, 1.4) 0.55
Total carotene (µg/d) 0.14 (0.0, 0.3) 0.12 0.13 (− 0.1, 0.3) 0.16 0.63 (− 0.2, 1.5) 0.14
Retinol (µg/d) 0.00 (− 0.2, 0.2) 0.99 – 0.01 (− 0.2, 0.2) 0.92 0.32 (− 0.6, 1.3) 0.51
Protein (%E/d) – 0.32 (− 0.5, − 0.2) < 0.01 – 0.34 (− 0.5, − 0.2) < 0.01 – 0.50 (− 1.5, 0.5) 0.34
LEPb (w/kg)
Vitamin C (mg/d) 2.14 (0.9, 3.3) < 0.01 2.32 (1.1, 3.6) < 0.01 – 1.06 (− 9.0, 6.9) 0.79
Vitamin E (mg/d) 0.66 (− 1.0, 2.3) 0.43 0.60 (− 1.2, 2.4) 0.51 – 0.76 (− 9.4, 7.8) 0.86
Total carotene (µg/d) 1.35 (0.1, 2.6) 0.03 1.67 (0.4, 2.9) < 0.01 – 6.68 (− 15.0, 1.6) 0.11
Retinol (µg/d) – 0.23 (− 1.5, 1.1) 0.73 – 0.39 (− 1.8, 1.0) 0.57 0.26 (− 8.8, 9.4) 0.95
Protein (%E/d) – 1.18 (− 2.3, 0.0) 0.04 – 1.18 (− 2.4, 0.0) 0.05 – 2.64 (− 8.6, 3.3) 0.38
FFMI (kg/m2)
Vitamin C (mg/d) 0.07 (0.0, 0.1) < 0.01 0.08 (0.03, 0.12) < 0.01 0.02 (− 0.3, 0.3) 0.91
Vitamin E (mg/d) 0.00 (− 0.05, 0.06) 0.85 – 0.01 (− 0.07, 0.05) 0.70 0.23 (− 0.1, 0.6) 0.17
Total carotene (µg/d) 0.06 (0.02, 0.1) < 0.01 0.05 (0.01, 0.1) 0.02 0.22 (− 0.03, 0.46) 0.09
Retinol (µg/d) – 0.03 (− 0.08, 0.02) 0.26 – 0.04 (− 0.1, 0.01) 0.11 0.13 (− 0.16, 0.43) 0.38
Protein (%E/d) 0.03 (− 0.01, 0.07) 0.15 0.03 (− 0.01, 0.1) 0.14 0.05 (− 0.23, 0.33) 0.73
FFM/BMI
Vitamin C (mg/d) 0.00 (− 0.0, 0.01) 0.08 0.01 (− 0.00, 0.01) 0.11 0.02 (− 0.01, 0.05) 0.26
Vitamin E (mg/d) 0.01 (0.0, 0.02) < 0.01 0.02 (0.01, 0.03) < 0.01 – 0.00 (− 0.04, 0.04) 0.96
Total carotene (µg/d) 0.00 (− 0.0, 0.0) 0.49 0.00 (− 0.00, 0.01) 0.54 0.02 (− 0.01, 0.05) 0.24
Retinol (µg/d) 0.00 (− 0.0, 0.01) 0.23 0.00 (− 0.00, 0.01) 0.24 0.01 (− 0.02, 0.05) 0.43
Protein (%E/d) – 0.02 (− 0.02, − 0.01) < 0.01 – 0.02 (− 0.02, − 0.01) < 0.01 – 0.02 (− 0.05, 0.02) 0.38

FFM fat free mass and LEP leg explosive power


a
Values are adjusted beta coefficients (95% CI) per quantile of intake, n = 2570 (Quintiles for all ages and < 65 years and tertiles for
those ≥ 65 year). Models were adjusted for age, physical activity, smoking status, energy intake and underreporting
b
Subset analysis n = 1914 (< 65 years n = 1794; ≥ 65 years n = 120). Means were adjusted for age, physical activity, smoking status, energy
intake, underreporting, menopausal status, hormone replacement therapy and height. Dietary variables are expressed per quintile for all partici-
pants and participants < 65 years and tertiles for participants ≥ 65 years

Comparison with Other Studies intake with FFM found positive associations but only after
2.6 years of follow-up [11].
The positive associations we found between higher intakes Circulating vitamin E was investigated in only three pre-
of vitamin C, carotenoids and vitamin E are, in the main, vious studies where plasma α-tocopherol was positively
supported by the few previous human studies that investi- associated with measures of grip and knee strength, perfor-
gated the relationships between intake or circulating con- mance or decline in physical function. However, two stud-
centrations of vitamin C, E or carotene and sarcopenic ies found no associations between intake of vitamin E and
indices in cohorts of older people [11–14]. Dietary vitamin either grip strength or function [12, 14, 17, 32]. Our
C was associated with measures of skeletal muscle func- finding of a positive relationship with dietary vitamin E
tion in the InCHIANTI Study, and in women only in the and certain indices of fat-free mass further supports the
UK Hertfordshire Cohort Study (HCS) [11–14]. Circulat- relevance of vitamin E to skeletal muscle health.
ing vitamin C and sarcopenic indices were only previously For carotenoids, two previous studies found associations
examined in elderly Japanese women where positive asso- with either circulating β or total carotene and greater
ciations were found between measures of physical function muscle strength or physical activity, with only one other
but not FFM [13]. The only study investigating vitamin C study find- ing positive associations between higher
intakes of carotene and strength or physical activity, in
women [12, 14–19]. Our
13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 33
Intakes…
Fig. 2 The relative associa-
tions of vitamin C, vitamin E, 0.8
retinol, carotene and protein 0.7
with leg explosive power in 0.6
1914 females aged 18–79 0.5 *
*
years, stratified by age1. Values 0.4
repre- sent the difference in 0.3
stand- 0.2
ardised values of leg explosive 0.1
power between participants in 0
Q5 vs Q1 (T3-T1 for > 65 years

SD
-0.1
sub-group) of intake with all -0.2
nutrients included in the model. -0.3
Values were also adjusted for -0.4 * *
age, physical activity, smoking -0.5
status, energy intake, underre- -0.6
porting, height, menopausal sta- -0.7
tus and use of hormone replace- -0.8
ment therapy. * P-trend < 0.05 -0.9
-1
All (n=2570) <65 y (n=2346) ≥65 y (n=224)
Vitamin C Vitamin E Total carotene Retinol Protein

Fig. 3 Foods that contributed 80


to at least 10% of vitamin C, 70
vitamin E, carotene and retinol
intakes in 2570 females aged 60
18–79 years1. Values are the 50
percentage contribution of 40
%

individual foods to total nutrient 30


intake. The main contributors to 20
vegetable intakes were peppers, 10
Brussels sprouts and broc-
0
coli for Vitamin C, avocado,
Fruits

Offal
Cakes and biscuits
Wholegrain cereals

Wholegrain cereals

Dairy
Vegetables

Vegetables

Vegetables
mushrooms and spinach for
Vitamin E and carrots and
spinach for carotene. For Vita-
min C the main contributors to
fruit intakes were strawberries,
oranges and grapefruit
due to LEP being a direct measure of lower limb strength
Vitamin C Vitamin E Carotene Retinol

findings extend the previous research by identifying that


the full range of carotenoids also have importance for
skeletal muscle health.
The lack of association with retinol intake and sarco-
penic indices found in our study contrasts with the only
other human study that found greater intakes of retinol
were significantly related to loss of appendicular lean mass
[11]. In recent studies, relating intakes of either vitamin C,
E,
or circulating vitamin E, to sarcopenia no associations
were found [33–35] although greater prevalence of frailty
was associated with either lower concentrations of
carotenoids, retinol or α-tocopherol, in a further three
studies [36–39].
The reason for the lack of association between the vita-
mins in our study and grip strength, compared with the
strong associations with LEP are not clear but may be
13
34 A. A. Welch et al.
Although we also found smaller associations with the
sarcopenic indices and protein intake than the vitamins in
our standardised analyses, other studies have found protein
and power which may be more sensitive to the effects of is important for skeletal muscle. However, there has been
diet [29]. variability in effectiveness of supplementation with protein
The lack of association between the vitamins and in intervention studies, and protein intake in our study was
CRP in our study may be due to the relatively high higher, 1.3 g protein/kg/d, than in other previous studies
intakes of vitamins, coupled with the relatively low blood which may explain our findings [10, 40]. However, our
concentra- tions of hsCRP in this population. findings relating to protein intake require further explora-
tion. Nevertheless, our findings suggest that consuming

13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 34
Intakes…
sufficient antioxidant vitamins in addition to protein is by a different food groups. Given the shortfall in intakes of
important.
Despite the few previous studies, to our knowledge, our
study is the first to investigate dietary intakes of vitamin C,
E, A (retinol), individual carotenoids and protein, as well
as circulating CRP with the full range of sarcopenic indices
in both younger and older women.

Physiological Mechanisms

There is clear mechanistic relevance of the antioxidant


vitamins on skeletal muscle health during aging which
also supports our observational findings. Supplementation
of lycopene, β-carotene or mixed antioxidant vitamins in
animal experiments either had positive effects on muscle
force or physical activity or attenuated oxidative stress or
loss of skeletal muscle [41]. Vitamin E also had protective
effects on exercise-induced oxidative damage in both
young and older adults [7], and vitamin C had protective
effects on oxidative biomarkers, inflammatory cytokines
and CRP [42]. Beyond roles as an antioxidant and anti-
inflammatory agent vitamin C is also integral to synthesis
of collagen and of carnitine which is important for the
metabolism of long- chain fatty acids during physical
activity [8].

Deficiency of Vitamins and Supplementation

Although the significant associations between intake of


vitamins C, E and carotenoids were found across the usual
range of consumption, the women in the lowest quintile of
vitamin C intake consumed less than the average require-
ment for vitamin C intake of 80 mg/d [43]. The prevalence
of low intakes of vitamin C, or circulating concentrations
of vitamin C indicative of scurvy, is high in vulnerable
older populations living in community and residential care
~ 40% [3, 44]. Moreover, consumption of 5 or more
portions of fruits and vegetables a day is only 19% of those
over the age of 75 years in the UK [45].
Whilst supraphysiological doses of vitamin C could be
used to rectify low dietary intakes in older vulnerable
groups these need to be used with caution due to the
detrimental, prooxidant, effects of long-term high doses
of vitamin C (> 500 mg per day) compared the
concentrations found natu- rally in foods [46]. There is also
debate surrounding poten- tial negative effects of
supraphysiological doses of vitamin C on skeletal muscle
function in athletes [47].
Our research has found that intake of antioxidant vita-
mins within the normal dietary range has important effects
on sarcopenic indices, in the whole cohort and stratified
analysis showed associations in younger women, provided

13
34 A. A. Welch et al.
the antioxidant vitamins it is important to encourage Trust (Grant WT081878MA). Medical Research Council, European
greater intakes of the foods that supply them; namely Union, the National Institute for Health Research (NIHR)-funded
BioResource,
vegetables, fruits and whole grain cereal foods, in
addition protein for maintaining skeletal muscle health in
populations of all ages. This is particularly important for
vulnerable older people in the community and in
residential care.
Strengths of this study include comprehensive meas-
urements of diet in a well characterised large cohort
which allowed us to examine for the first time
associations between an extensive range of sarcopenic
indices, including DXA-measured FFM, LEP and grip
strength and a range of antioxidant vitamins, including
all the carotenoids. We were also able to undertake a
comparative analysis of the different nutrients allowing
us to examine the independ- ent associations with intake
of each vitamin and protein. Limitations include the small
number of women aged over 65 an imbalance between
younger and older women in our cohort which future
studies should address. In addition, this study was cross-
sectional which limits inference for causa- tion. We also
did not account for the nutrients supplied by food
supplements, however, our analyses indicate the impor-
tance of intake of vitamins supplied by diet alone. A
further limitation is that we did not have directly
measured physical activity, however, we used validated
questionnaires, which although less precise than objective
measures, do distinguish across the range of activity
levels in individuals [28].
Although we were able to determine what dietary intakes
are relevant for skeletal muscle health we lacked data on
dietary biomarkers meaning we were unable to confirm
our findings with blood or urinary levels.

Conclusion

For the first time in a free-living population, our research


shows that higher dietary intake of the antioxidant
vitamins, particularly vitamin C, could be protective for
both loss of skeletal muscle mass and power during
aging, and have rel- evance for treatment and prevention
of frailty and sarcopenia in women. It was notable that the
observed associations were identified in women younger
than 65 years. Our findings provide further
encouragement for intervention trials as well as for
following the healthy eating guidelines for protection of
skeletal muscle mass and power in people of all ages.

Author Contributions Concept and design of study AAW, AJ, EK.


Statistical analysis AJ. Drafting of manuscript AAW, AJ. All
authors commented on the manuscript and contributed to the study.

Funding This study was funded by PhD funding from the


University of East Anglia. TwinsUK is funded by the Wellcome
13
Cross-Sectional Associations Between Dietary Antioxidant Vitamins C, E and Carotenoid 34
Intakes…
Clinical Research Facility, and Biomedical Research Centre based
at Guy’s and St Thomas’ NHS Foundation Trust in partnership with 9. Cruz-Jentoft AJ, Woo J (2019) Nutritional interventions to prevent
King’s College London. and treat frailty. Curr Opin Clin Nutr Metab Care. https://doi.
org/10.1097/MCO.0000000000000556
10. Thomas DK, Quinn MA, Saunders DH, Greig CA (2016) Pro-
Compliance with Ethical Standards tein supplementation does not significantly augment the effects
of resistance exercise training in older adults: a systematic
Conflict of interest Ailsa Welch, Amy Jennings, Eirini Kelaiditi, Jane review. J Am Med Dir Assoc 17(10):959e951–959. https://doi.
Skinner and Claire Steves declare no conflicts of interest. org/10.1016/j.jamda.2016.07.002
11. Scott D, Blizzard L, Fell J, Giles G, Jones G (2010) Associations
Human and Animal Rights and Informed Consent This study was between dietary nutrient intake and muscle mass and strength
approved by the St Thomas’s Hospital Research Ethics Committee in community-dwelling older adults: the Tasmanian Older Adult
and all participants provided written informed consent. Cohort Study. J Am Geriatr Soc 58(11):2129–2134. https://doi.
org/10.1111/j.1532-5415.2010.03147.x
12. Martin H, Aihie Sayer A, Jameson K, Syddall H, Dennison EM,
Open Access This article is licensed under a Creative Commons Attri- Cooper C, Robinson S (2011) Does diet influence physical per-
bution 4.0 International License, which permits use, sharing, adapta- formance in community-dwelling older people? Findings from
tion, distribution and reproduction in any medium or format, as long the Hertfordshire Cohort Study. Age Ageing 40(2):181–186.
as you give appropriate credit to the original author(s) and the source, https:// doi.org/10.1093/ageing/afq175
provide a link to the Creative Commons licence, and indicate if 13. Saito K, Yokoyama T, Yoshida H, Kim H, Shimada H, Yoshida
changes were made. The images or other third party material in this Y, Iwasa H, Shimizu Y, Kondo Y, Handa S, Maruyama N, Ish-
article are included in the article’s Creative Commons licence, unless igami A, Suzuki T (2012) A significant relationship between
indicated otherwise in a credit line to the material. If material is not plasma vitamin C concentration and physical performance
included in the article’s Creative Commons licence and your intended among Japanese elderly women. J Gerontol A Biol Sci Med Sci
use is not permitted by statutory regulation or exceeds the permitted 67(3):295–301. https://doi.org/10.1093/gerona/glr174
use, you will need to obtain permission directly from the copyright 14. Robinson SM, Jameson KA, Batelaan SF, Martin HJ, Syddall
holder.To view a copy of this licence, visit HE, Dennison EM, Cooper C, Sayer AA (2008) Diet and its
https://creativecommons.org/licenses/by/4.0/. rela- tionship with grip strength in community-dwelling older
men and women: the Hertfordshire cohort study. J Am Geriatr
Soc 56(1):84–90. https://doi.org/10.1111/j.1532-
5415.2007.01478.x
15. Cesari M, Pahor M, Bartali B, Cherubini A, Penninx BW, Wil-
References liams GR, Atkinson H, Martin A, Guralnik JM, Ferrucci L
(2004) Antioxidants and physical performance in elderly per-
1. Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyere O, Ceder- sons: the Invecchiare in Chianti (InCHIANTI) study. Am J Clin
holm T, Cooper C, Landi F, Rolland Y, Sayer AA, Schneider Nutr 79(2):289–294
SM, Sieber CC, Topinkova E, Vandewoude M, Visser M, 16. Lauretani F, Semba RD, Bandinelli S, Dayhoff-Brannigan M,
Zamboni M, Writing Group for the European Working Group on Giacomini V, Corsi AM, Guralnik JM, Ferrucci L (2008) Low
Sarcopenia in Older P, the Extended Group for E (2019) plasma carotenoids and skeletal muscle strength decline over 6
Sarcopenia: revised European consensus on definition and years. J Gerontol A Biol Sci Med Sci 63(4):376–383
diagnosis. Age Ageing 48(1):16–31. 17. Semba RD, Blaum C, Guralnik JM, Moncrief DT, Ricks MO,
https://doi.org/10.1093/ageing/afy169 Fried LP (2003) Carotenoid and vitamin E status are associated
2. Landi F, Camprubi-Robles M, Bear DE, Cederholm T, Malafa- with indicators of sarcopenia among older women living in the
rina V, Welch AA, Cruz-Jentoft AJ (2018) Muscle loss: the new community. Aging Clin Exp Res 15(6):482–487
malnutrition challenge in clinical practice. Clin Nutr. https://doi. 18. Semba RD, Varadhan R, Bartali B, Ferrucci L, Ricks MO,
org/10.1016/j.clnu.2018.11.021 Blaum C, Fried LP (2007) Low serum carotenoids and devel-
3. Bunn D, Hooper L, Welch A (2018) Dehydration and opment of severe walking disability among older women living
malnutrition in residential care: recommendations for strategies in the community: the women’s health and aging study I. Age
for improving practice derived from a scoping review of existing Ageing 36(1):62–67. https://doi.org/10.1093/ageing/afl122
policies and guidelines. Geriatrics 3(4):77 19. Alipanah N, Varadhan R, Sun K, Ferrucci L, Fried LP, Semba
4. Cesari M, Araujo de Carvalho I, Amuthavalli Thiyagarajan J, RD (2009) Low serum carotenoids are associated with a
Cooper C, Martin FC, Reginster JY, Vellas B, Beard JR (2018) decline in walking speed in older women. J Nutr Health Aging
Evidence for the domains supporting the construct of intrinsic 13(3):170–175
capacity. J Gerontol A Biol Sci Med Sci 73(12):1653–1660. 20. Moayyeri A, Hammond CJ, Valdes AM, Spector TD (2013)
https Cohort profile: TwinsUK and healthy ageing twin study. Int J
://doi.org/10.1093/gerona/gly011 Epidemiol 42(1):76–85. https://doi.org/10.1093/ije/dyr207
5. Dalle S, Rossmeislova L, Koppo K (2017) The role of 21. McCance RA, Widdowson EM, Holland B, Welch AA, Buss
inflamma- tion in age-related sarcopenia. Front Physiol 8:1045. DH, Great Britain Ministry of Agriculture F, Food (1991)
https://doi. org/10.3389/fphys.2017.01045 McCance and Widdowson’s the composition of foods, 5th rev
6. Chapman MA, Meza R, Lieber RL (2016) Skeletal muscle fibro- & extended. Royal Society of Chemistry, London
blasts in health and disease. Differentiation 92(3):108–115. https 22. Hayhoe RPG, Lentjes MAH, Mulligan AA, Luben RN, Khaw
://doi.org/10.1016/j.diff.2016.05.007 KT, Welch AA (2017) Carotenoid dietary intakes and plasma
7. Chung E, Mo H, Wang S, Zu Y, Elfakhani M, Rios SR, Chyu concentrations are associated with heel bone ultrasound attenu-
MC, Yang RS, Shen CL (2018) Potential roles of vitamin E in ation and osteoporotic fracture risk in the European Prospective
age-related changes in skeletal muscle health. Nutr Res 49:23– Investigation into Cancer and Nutrition (EPIC)-Norfolk cohort.
36. https://doi.org/10.1016/j.nutres.2017.09.005 Br J Nutr 117(10):1439–1453. https://doi.org/10.1017/S0007
8. Rebouche CJ (1152S) Ascorbic acid and carnitine biosynthe- 114517001180
sis. Am J Clin Nutr 54(6 Suppl):1147S–1152S. https://doi.
org/10.1093/ajcn/54.6.1147s
13
34 A. A. Welch et al.

23. Otten JJ, Hellwig JP, Meyers LD (2006) DRI, dietary reference
intakes: the essential guide to nutrient requirements. National older women living in the community. J Gerontol Ser A Biol Sci
Academies Press, Washington, D.C. Med Sci 61(6):594–599
24. Arden NK, Spector TD (1997) Genetic influences on mus- 37. Michelon E, Blaum C, Semba RD, Xue QL, Ricks MO, Fried LP
cle strength, lean body mass, and bone mineral density: a (2006) Vitamin and carotenoid status in older women: associa-
twin study. J Bone Miner Res 12(12):2076–2081. https://doi. tions with the frailty syndrome. J Gerontol A Biol Sci Med Sci
org/10.1359/jbmr.1997.12.12.2076 61(6):600–607
25. Bassey EJ, Short AH (1990) A new method for measuring 38. Ble A, Cherubini A, Volpato S, Bartali B, Walston JD,
power output in a single leg extension: feasibility, reliability and Windham BG, Bandinelli S, Lauretani F, Guralnik JM, Ferrucci
valid- ity. Eur J Appl Physiol Occupational Physiol 60(5):385– L (2006) Lower plasma vitamin E levels are associated with the
390 frailty syndrome: the InCHIANTI study. J Gerontol A Biol Sci
26. MacGregor AJ, Gallimore JR, Spector TD, Pepys MB (2004) Med Sci 61(3):278–283
Genetic effects on baseline values of C-reactive protein and 39. Pilleron S, Weber D, Peres K, Colpo M, Gomez-Cabrero D, Stu-
serum amyloid a protein: a comparison of monozygotic and etz W, Dartigues JF, Ferrucci L, Bandinelli S, Garcia-Garcia FJ,
dizygotic twins. Clin Chem 50(1):130–134. https ://doi. Grune T, Feart C, Initiative F (2018) Patterns of circulating fat-
org/10.1373/clinchem.2003.028258 soluble vitamins and carotenoids and risk of frailty in four Euro-
27. Cherkas LF, Hunkin JL, Kato BS, Richards JB, Gardner JP, pean cohorts of older adults. Eur J Nutr. https://doi.org/10.1007/
Surdulescu GL, Kimura M, Lu X, Spector TD, Aviv A (2008) s00394-017-1602-0
The association between physical activity in leisure time and 40. Houston DK, Nicklas BJ, Ding J, Harris TB, Tylavsky FA,
leukocyte telomere length. Arch Intern Med 168(2):154–158. New- man AB, Lee JS, Sahyoun NR, Visser M, Kritchevsky SB
https://doi.org/10.1001/archinternmed.2007.39 (2008) Dietary protein intake is associated with lean mass
28. Wareham NJ, Jakes RW, Rennie KL, Schuit J, Mitchell J, Hen- change in older, community-dwelling adults: the Health, Aging,
nings S, Day NE (2003) Validity and repeatability of a simple and Body Composition (Health ABC) Study. Am J Clin Nutr
index derived from the short physical activity questionnaire used 87(1):150–155
in the European Prospective Investigation into Cancer and Nutri- 41. van Dijk M, Dijk FJ, Hartog A, van Norren K, Verlaan S, van
tion (EPIC) study. Public Health Nutr 6(04):407–413. Helvoort A, Jaspers RT, Luiking Y (2018) Reduced dietary
https://doi. org/10.1079/PHN2002439 intake of micronutrients with antioxidant properties negatively
29. Mitchell WK, Williams J, Atherton P, Larvin M, Lund J, Narici impacts muscle health in aged mice. J Cachexia Sarcopenia
M (2012) Sarcopenia, dynapenia, and the impact of advancing Muscle 9(1):146–159. https://doi.org/10.1002/jcsm.12237
age on human skeletal muscle size and strength; a quantitative 42. Block G, Jensen CD, Dalvi TB, Norkus EP, Hudes M, Crawford
review. Front Physiol 3:260. PB, Holland N, Fung EB, Schumacher L, Harmatz P (2009)
https://doi.org/10.3389/fphys.2012.00260 Vitamin C treatment reduces elevated C-reactive protein. Free
30. Cesari M, Pahor M (2008) Target population for clinical trials Radic Biol Med 46(1):70–77.
on sarcopenia. J Nutr Health Aging 12(7):470–478. https://doi. https://doi.org/10.1016/j.freeradbio med.2008.09.030
org/10.1007/bf02982708 43. EFSA (2013) Scientific opinion on dietary reference values for
31. Janssen I, Heymsfield SB, Wang ZM, Ross R (2000) Skeletal vitamin C. EFSA J 11(11):3418
muscle mass and distribution in 468 men and women aged 18– 44. Schleicher RL, Carroll MD, Ford ES, Lacher DA (2009) Serum
88 year. J Appl Physiol (1985) 89(1):81–88. vitamin C and the prevalence of vitamin C deficiency in the
https://doi.org/10.1152/ jappl.2000.89.1.81 United States: 2003–2004 National Health and Nutrition Exami-
32. Bartali B, Frongillo EA, Guralnik JM, Stipanuk MH, Allore HG, nation Survey (NHANES). Am J Clin Nutr 90(5):1252–1263.
Cherubini A, Bandinelli S, Ferrucci L, Gill TM (2008) Serum https://doi.org/10.3945/ajcn.2008.27016
micronutrient concentrations and decline in physical func- 45. England PH (2019) National diet and nutrition survey. https://
tion among older persons. JAMA 299(3):308–315. https://doi. www.gov.uk/government/statistics/national-diet-and-nutrition-
org/10.1001/jama.299.3.308 survey-results-from-years-1-to-4-combined-of-the-rolling-progr
33. Chaput JP, Lord C, Cloutier M, Aubertin Leheudre M, Goulet amme-for-2008-and-2009-to-2011-and-2012. Accessed 24 April
ED, Rousseau S, Khalil A, Dionne IJ (2007) Relationship 2019
between antioxidant intakes and class I sarcopenia in elderly 46. Osiecki M, Ghanavi P, Atkinson K, Nielsen LK, Doran MR
men and women. J Nutr Health Aging 11(4):363–369 (2010) The ascorbic acid paradox. Biochem Biophys Res Com-
34. Verlaan S, Aspray TJ, Bauer JM, Cederholm T, Hemsworth J, mun 400(4):466–470. https://doi.org/10.1016/j.bbrc.2010.08.052
Hill TR, McPhee JS, Piasecki M, Seal C, Sieber CC, Ter Borg 47. Mason SA, Morrison D, McConell GK, Wadley GD (2016)
S, Wijers SL, Brandt K (2017) Nutritional status, body com- Mus- cle redox signalling pathways in exercise. Role of
position, and quality of life in community-dwelling sarcopenic antioxidants. Free Radic Biol Med 98:29–45.
and non-sarcopenic older adults: a case-control study. Clin Nutr https://doi.org/10.1016/j.freer adbiomed.2016.02.022
36(1):267–274. https://doi.org/10.1016/j.clnu.2015.11.013
35. Ter Borg S, de Groot LC, Mijnarends DM, de Vries JH, Verlaan Publisher’s Note Springer Nature remains neutral with regard to
S, Meijboom S, Luiking YC, Schols JM (2016) Differences in jurisdictional claims in published maps and institutional affiliations.
nutrient intake and biochemical nutrient status between sarco-
penic and nonsarcopenic older adults-results from the maastricht
sarcopenia study. J Am Med Dir Assoc.
https://doi.org/10.1016/j. jamda.2015.12.015
36. Semba RD, Bartali B, Zhou J, Blaum C, Ko CW, Fried LP
(2006) Low serum micronutrient concentrations predict frailty
among

13
REVIEW
published: 17 April 2019
doi: 10.3389/fnut.2019.00048

Metabolism of Dietary and Microbial


Vitamin B Family in the Regulation of
Host Immunity
Ken Yoshii 1,2, Koji Hosomi 1, Kento Sawane 1,3,4 and Jun Kunisawa 1,2,3,5,6,7*
1
Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System,
National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan, 2 Graduate School of Medicine, Osaka
University, Osaka, Japan, 3 Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan, 4 Innovation
Center, Nippon Flour Mills Co., Ltd., Atsugi, Japan, 5 Graduate School of Dentistry, Osaka University, Osaka, Japan,
6
Department of Microbiology and Immunology, Graduate School of Medicine, Kobe University, Hyogo, Japan, 7 Division of
Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science,
The University of Tokyo, Tokyo, Japan

Vitamins are micronutrients that have physiological effects on various biological responses,
including host immunity. Therefore, vitamin deficiency leads to increased risk of developing
infectious, allergic, and inflammatory diseases. Since B vitamins are synthesized by plants,
yeasts, and bacteria, but not by mammals, mammals must acquire B vitamins from dietary or
microbial sources, such as the intestinal microbiota. Similarly, some intestinal bacteria are unable
to synthesize B vitamins and must acquire them from the host diet or from other intestinal
Edited by: bacteria for their growth and survival. This suggests that the composition and function of the
Carlotta De Filippo, intestinal microbiota may affect host B vitamin usage and, by extension, host immunity.
Italian National Research Council
(CNR), Italy
Here, we review the immunological functions of B vitamins and their metabolism by
Reviewed by:
intestinal bacteria with respect to the control of host immunity.
Chiara Devirgiliis,
Keywords: absorption, gut microbiota, intestinal immunity, nutrition, vitamin
Council for Agricultural Research and
Economics, Italy
Williams Turpin,
University of Toronto, Canada INTRODUCTION
*Correspondence:
The gut is continuously exposed both to toxic (e.g., pathogenic microorganisms) and beneficial
Jun Kunisawa
kunisawa@nibiohn.go.jp
(e.g., dietary components, commensal bacteria) compounds and microorganisms; therefore, the
intestinal immune system must maintain a healthy balance between active and suppressive immune
Specialty section:
responses. This balance is controlled not only by host immune factors such as cytokines but also by a
This article was submitted to variety of environmental factors such as dietary components and the composition of the commensal
Nutrition and Microbes, bacteria. Furthermore, several lines of evidence have demonstrated that immune homeostasis
a section of the journal in the intestine is related not only to intestinal health but also to the health of the whole body
Frontiers in Nutrition (1– 3). Therefore, immune regulation by environmental factors is attracting attention as a means
Received: 05 December 2018 of maintaining immunological health and preventing many diseases.
Accepted: 01 April 2019 Nutrients are essential for the development, maintenance, and function of the host immune
Published: 17 April 2019 system (4, 5). Vitamins are essential micronutrients that are synthesized by bacteria, yeasts, and
Citation: plants, but not by mammals. Therefore, mammals must obtain vitamins from the diet or rely on
Yoshii K, Hosomi K, Sawane K and their synthesis by commensal bacteria in the gastrointestinal tract. Some vitamins are water-soluble
Kunisawa J (2019) Metabolism of (e.g., vitamin B family and vitamin C), whereas others are fat-soluble (e.g., vitamins A, D, E, and
Dietary and Microbial Vitamin B Family
K). Water-soluble vitamins are not stored by the body and any excess is excreted in the urine;
in the Regulation of Host Immunity.
therefore, it is important to consume a diet containing the necessary amounts of these
Front. Nutr. 6:48.
doi: 10.3389/fnut.2019.00048 vitamins. Vitamin

Frontiers in Nutrition | www.frontiersin.org 1 April 2019 | Volume 6 | Article 48


Yoshii et al. Vitamin B Family and Host Immunity

deficiency associated with insufficient dietary intake occurs not induced by vitamin B1
only in developing countries but also in developed countries as
a result of increased use of unbalanced diet (6).
In addition to the diet, the commensal bacteria are recognized
as important players in the control of host health (7–9). From
the point of view of vitamins, commensal bacteria are both
providers and consumers of B vitamins and vitamin K.
Although dietary B vitamins are generally absorbed through
the small intestine, bacterial B vitamins are produced and
absorbed mainly through the colon (10, 11), indicating that
dietary and gut microbiota- derived B vitamins are possibly
handled differently by the human body. B vitamins are
important cofactors and coenzymes in several metabolic
pathways, and it has been reported recently that B vitamins
also play important roles in the maintenance of immune
homeostasis (12, 13). Thus, both dietary components and the
gut microbiota modulate host immune function via B
vitamins. Here, we review the metabolism and function of
dietary and gut microbiota-derived B vitamins in the control of
host immunity.

VITAMIN B1
Vitamin B1 (thiamine) is a cofactor for several enzymes,
including pyruvate dehydrogenase and α-ketoglutarate
dehydrogenase, which are both involved in the tricarboxylic
acid (TCA) cycle (14, 15). World Health Organization
(WHO)/Food and Agriculture Organization (FAO) recommend
a daily vitamin B1 intake of 1.1–1.2 mg for adult (16). Vitamin
B1 deficiency causes lethargy and, if left untreated, can
develop into beriberi, a disease that affects the peripheral
nervous system and cardiovascular system. Accumulating
evidence suggests that energy metabolism—particularly the
balance between glycolysis and the TCA cycle—is associated
with the functional control of immune cells, in what is now
referred to as immunometabolism (17).
Immunometabolism is well studied in T cells and
macrophages; quiescent or regulatory-type cells (e.g., naive
T cells, Treg cells, and M2 macrophages) use the TCA cycle for
energy generation, whereas activated or pro-inflammatory
cells (e.g., Th1, Th2, Th17, and M1 macrophages) use
glycolysis (18, 19).
Previously, we examined B cell immunometabolism in the
intestine. In the intestine, naïve immunoglobulin (Ig) M + B cells
differentiate into IgA+ B cells in Peyer’s patches (PPs) by class
switching, and then IgA+ B cells differentiate into IgA-
producing plasma cells in the intestinal lamina propria (20).
Naïve B cells
in PPs preferentially use a vitamin B1-dependent TCA cycle for
the generation of ATP. However, once the B cells differentiate
into IgA-producing plasma cells, they switch to using glycolysis
for the generation of ATP and shift to a catabolic pathway for
the production of IgA antibody (Figure 1). Consistent with the
importance of vitamin B1 in the maintenance of the TCA cycle,
mice fed a vitamin B1-deficient diet show impaired
maintenance of naïve B cells in PPs, with little effect on IgA-
producing plasma cells. Since PPs are the primary sites of
induction of antigen- specific IgA responses, PP regression
Frontiers in Nutrition | www.frontiersin.org 2 April 2019 | Volume 6 | Article 48
Yoshii et al. Vitamin B Family and Host Immunity

deficiency leads to decreased IgA antibody responses to oral


vaccines (21).
Vitamin B1 is found in high concentrations as thiamine
pyrophosphate (TPP) in meat (particularly pork and chicken);
eggs; cereal sprouts and rice bran; and beans. Dietary TPP
is hydrolyzed by alkaline phosphatase and converted to free
thiamine in the small intestine (22). Free thiamine is absorbed
by the intestinal epithelium via thiamine transporters (e.g.,
THTR-1, THTR-2) and transported to the blood for distribution
throughout the body (11). Free thiamine is converted back to TPP
and is used for energy metabolism in the TCA cycle.
Various types of intestinal bacteria, mostly in the colon,
also produce vitamin B1 as both free thiamine and TPP
(11, 23). In the colon, free bacterial thiamine is absorbed
mainly by thiamine transporters, transported to the blood,
and distributed throughout the body; this mechanism is
similar to how free dietary thiamine is taken up in the small
intestine. However, unlike in the small intestine, TPP produced
by the gut microbiota is not converted to free thiamine,
because alkaline phosphatase is not secreted in the colon
(24). Instead, TPP is absorbed directly by the colon via TPP
transporters (e.g., TPPT-1) that are highly expressed on the
apical membrane of the colon (25). The absorbed TPP
enters the mitochondria via MTPP-1, a TPP transporter that
is expressed in the mitochondrial inner membrane and is used
as a cofactor for ATP generation (26). This suggests that
bacterial TPP is important for energy generation in the colon.
Thus, dietary and bacterial vitamin B1 appears to have
different roles in the host.
The vitamin B1 structure consists of a thiazole moiety
linked to a pyrimidine moiety. Bacteria obtain the thiazole
moiety from glycine or tyrosine and 1-deoxy-D-xylulose-5-
phosphate, and plants and yeasts synthesize it from glycine
and 2-pentulose (27– 30). In both bacteria and plants, the
pyrimidine moiety is derived from 5-aminoimidazole
ribonucleotide, an intermediate in the purine pathway (29).
Metagenomic analyses of the human gut microbiota predict
that Bacteroides fragilis and Prevotella copri (phylum
Bacteroidetes); Clostridium difficile, some Lactobacillus spp.,
and Ruminococcus lactaris (Firmicutes); Bifidobacterium spp.
(Actinobacteria); and Fusobacterium varium are vitamin B1
producers (Table 1) (10, 46), implying that many intestinal
bacteria possess a complete vitamin B1 synthesis pathway, which
includes pathways for the synthesis of thiazole and
pyrimidine. Indeed, Lactobacillus casei produces thiamine
during the production of fermented milk drinks (31), and
Bifidobacterium infantis and B. bifidum produce thiamine in
culture supernatant (32). However, Faecalibacterium spp.
(Firmicutes) lack a vitamin B1 synthesis pathway even though
they require vitamin B1 for their growth (10). Therefore, these
bacteria must obtain their vitamin B1 from other bacteria or
from the host diet via a thiamine transporter, suggesting that
there is competition for vitamin B1 between the host and
certain intestinal bacteria.

VITAMIN B2
Vitamin B2 (riboflavin) and its active forms (flavin adenine
dinucleotide [FAD] and flavin mononucleotide [FMN]) are

Frontiers in Nutrition | www.frontiersin.org 3 April 2019 | Volume 6 | Article 48


Yoshii et al. Vitamin B Family and Host Immunity

cofactors for enzymatic reactions in the TCA cycle and in fatty L. fermentum, and Ruminococcus lactaris (Firmicutes) express
acid oxidization (also known as β-oxidization) (15). WHO/FAO factors essential for vitamin B2 synthesis, suggesting that these
recommends a daily
FIGURE 1 | Vitamin vitamin
B1 and B2 intake
B2-mediated of 1.0–1.3inmg
immunometabolism fordifferentiation
B cell adults inbacteria
the intestine.are an B1
Vitamin important source
acts as a cofactor of vitamin
for enzymes such as B2 in the
pyruvate large and α-ketogl
dehydrogenase
(16). Vitamin B2 deficiency suppresses the activity of acyl- intestine (Table 1). In contrast, Bifidobacterium spp., and
CoA dehydrogenases involved in the oxidation of fatty acids to Collinsella spp. (Actinobacteria) lack a vitamin B2 pathway.
generate acetyl-CoA, which is used by mitochondria to produce Supplementation of fermented soymilk containing
ATP via the TCA cycle. Fatty acid oxidization is involved Lactobacillus plantarum with riboflavin deficient diet has been
in the activation, differentiation, and proliferation of immune shown to promote vitamin B2 production and prevent vitamin
cells through the generation of acetyl-CoA and its entry into B2 deficiency in mice (35). L. fermentum isolated from
TCA cycle (47). This suggests that vitamin B2 is associated sourdough can synthesize riboflavin in vitro (36). Furthermore,
with the control of differentiation and function of immune recent evidence indicates that some species in Bacteroidetes
cells through regulation of fatty acid oxidization (Figure 1); phylum produce more riboflavin than do Actinobacteria and
however, the immunological roles of vitamin B2 in the control Firmicutes phyla (55). However, Actinobacteria and
of host immunity remain to be investigated. In addition to Firmicutes phyla still express riboflavin transporter and the
energy generation, vitamin B2 is associated with reactive enzymes necessary for FAD and FMN generation from free
oxygen species (ROS) generation in immune cells through the riboflavin (i.e., FAD synthases and flavin kinases) (10, 56),
priming of NADPH oxidase 2 (48); ROS are important effector suggesting that all bacteria, including those that are unable to
and signaling molecules in inflammation and immunity. synthesize vitamin B2 themselves, require FAD and FMN for
Vitamin B2 is found at high levels in leafy green vegetables, their growth and survival. Thus, as with vitamin B1, there is
liver, and eggs. Dietary vitamin B2 exists as FAD or FMN and is likely competition for riboflavin between the host and the
converted to free riboflavin by FAD pyrophosphatase and FMN commensal bacteria.
phosphatase in the small intestine (49, 50). Free riboflavin is In addition to being able to produce vitamin B2, some
absorbed via riboflavin transporter expressed on the bacteria (e.g., commensals such as Lactobacillus acidophilus
epithelium of the small intestine and is then released into the and pathogens such as Mycobacterium tuberculosis and
blood. In the blood, free riboflavin is converted back to FAD or Salmonella typhimurium) produce the vitamin B2 intermediate
FMN and distributed throughout the body (51–53). (57–59), 6- hydroxymethyl-8-D-ribityllumazine (60, 61). 6-
Bacterial vitamin B2 is synthesized from guanosine Hydroxymethyl- 8-D-ribityllumazine binds to major
triphosphate (GTP) and D-ribulose 5-phosphate (54). Bacterial histocompatibility complex class I-related gene protein (MR1)
vitamin B2 exists as free riboflavin, which is directly absorbed on antigen-presenting cells; this causes mucosal-associated
in the large intestine, converted to FAD or FMN, and invariant T (MAIT) cells, an abundant population of innate-
distributed throughout the body as described above (23). A like T cells, to produce cytokines such as interferon gamma
metagenome analysis of the human gut microbiota by and interleukin (IL) 17, which contribute to host defense
Magnúsdóttir et al. against pathogens (Figure 2) (62). It is thought that
(10) has predicted that Bacteroides fragilis and Prevotella copri stimulation by commensal bacteria contributes to the
(Bacteroidetes); Clostridium difficile, Lactobacillus plantarum, development and activation of MAIT cells for immunological
surveillance against pathogens. MAIT cells

Frontiers in Nutrition | www.frontiersin.org 3 April 2019 | Volume 6 | Article 48


Yoshii et al. Vitamin B Family and Host Immunity

TABLE 1 | Vitamin B family producing bacteria.


also produce inflammatory cytokines and have tissue-homing
Vitamins Forms Bacteria References properties, suggesting that these cells are also involved in the
development of autoimmune and inflammatory diseases (63).
B1 Thiamin Bacteroides fragilis (10, 31–33)
pyrophosphate Prevotella copri
(TPP) Clostridium difficile VITAMIN B3
Lactobacillus casei
Lactobacillus curvatus Vitamin B3 (niacin) is generally known as nicotinic acid and
Lactobacillus plantarum nicotinamide. These compounds are precursors of
Ruminococcus lactaris
Bifidobacterium infantis
nicotinamide adenine dinucleotide (NAD), a coenzyme in the
Bifidobacterium bifidum cellular redox reaction with a central role in aerobic
Fusobacterium varium respiration. WHO/FAO recommends a daily vitamin B3 intake
B2 Flavin adenine Bacteroides fragilis (10, 34–36) of 11–12 mg for adults (16).
dinucleotide (FAD) Prevotella copri
Flavin Vitamin B3 is also a ligand of GPR109a, a G-protein coupled
Clostridium difficile
mononucleotide Lactobacillus plantarum
receptor expressed on several types of cells, including immune
(FMN) Lactobacillus fermentum cells (64). Vitamin B3–GPR109a signaling induces
Ruminococcus lactaris differentiation of regulatory T cells and suppresses colitis in a
B3 Nicotinic acid Bacteroides fragilis (10, 32) GPR109a- dependent manner (65). Vitamin B3 also inhibits the
Nicotinamide Prevotella copri
production of the pro-inflammatory cytokines IL-1, IL-6, and
Ruminococcus lactaris
Clostridium difficile tumor necrosis factor alpha (TNF-α) by macrophages and
Bifidobacterium infantis monocytes (Figure 3) (66). Thus, vitamin B3 has anti-
Helicobacter pylori inflammatory properties by modulating host immune cells
Fusobacterium varium
B5 Free pantothenic
and playing an important role in the maintenance of
Bacteroides fragilis (10)
acid Prevotella copri
immunological homeostasis. Indeed, in humans, vitamin B3
Ruminococcus lactaris deficiency causes pellagra, which is a disease characterized by
Ruminococcus torques intestinal inflammation, diarrhea, dermatitis, and dementia
Salmonella enterica (67).
Helicobacter pylori
B6 Pyridoxal
Unlike the other B vitamins, vitamin B3 can be generated
Bacteroides fragilis (10, 32)
phosphate (PLP) Prevotella copri by mammals via an endogenous enzymatic pathway from
Bifidobacterium longum tryptophan and is stored in the liver, although it is also
Collinsella aerofaciens obtained from the diet (68). Animal-based foods such as fish
Helicobacter pylori and meat contain vitamin B3 as nicotinamide, and plant-based
foods such as beans contain vitamin B3 as nicotinic acid. Both
nicotinamide
B7 Free biotin Bacteroides fragilis (10, 37) and nicotinic acid are directly absorbed through the small
Lactobacillus helveticus
intestine, where nicotinic acid is converted to nicotinamide.
Fusobacterium varium
Campylobacter coli Vitamin B3 is also synthesized from tryptophan by intestinal
B9 Tetrahydrofolate Bacteroides fragilis (10, 38–41) bacteria (69, 70). Bacteroides fragilis and Prevotella copri
(THF) Prevotella copri (Bacteroidetes); Ruminococcus lactaris, Clostridium difficile
Clostridium difficile (Firmicutes); Bifidobacterium infantis (Actinobacteria);
Lactobacillus plantarum
Helicobacter pylori (Proteobacteria); and Fusobacterium varium
Lactobacillus delbrueckii
ssp. bulgaricus (Fusobacteria) possess a vitamin B3 biosynthesis pathway
Lactobacillus reuteri (Table 1) (10, 71). Thus, many intestinal bacteria from various
Streptococcus thermophilus genera can produce vitamin B3, suggesting that both dietary
Bifidobacterium and commensal bacteria-derived vitamin B3 are important for
pseudocatenulatum
host immunity.
Bifidobacterium
adolescentis
Fusobacterium varium
Salmonella enterica VITAMIN B5
B12 Adenosylcobalamin Bacteroides fragilis (10, 32, 33,
Fusobacterium
Prevotella copri 42–45)
Clostridium difficile
Faecalibacterium prausnitzii
Ruminococcus lactaris
Propionibacterium
freudenreichii
Lactobacillus plantarum
Lactobacillus coryniformis
Lactobacillus s reuteri
Bifidobacterium animalis
Bifidobacterium infantis
Bifidobacterium longum
Frontiers in Nutrition | www.frontiersin.org 4 April 2019 | Volume 6 | Article 48
Yoshii et al. Vitamin B Family and Host Immunity

Vitamin B5 (pantothenic acid) is a precursor of


coenzyme A (CoA), which is an essential
cofactor for the TCA cycle and fatty acid
oxidation (72). WHO/FAO recommends a daily
vitamin B5 intake of 5.0 mg for adults (16). Like
vitamins B1 and B2, vitamin B5 is involved in
the control of host immunity via energy
generation by immune cells. Vitamin B5 deficiency
causes immune diseases such as dermatitis, as
well as non-immune- related conditions such as
fatigue and insomnia (73). In a randomized,
double-blind, placebo-controlled study in
adults, dietary supplementation with vitamin
B5 was shown to improve facial acne (74),
suggesting that epithelial barrier function

Frontiers in Nutrition | www.frontiersin.org 5 April 2019 | Volume 6 | Article 48


Yoshii et al. Vitamin B Family and Host Immunity

FIGURE 2 | Regulation of MAIT cells by microbial metabolites derived from vitamin B2 and B9. Commensal bacteria/pathogens produce the vitamin B2 metabolite 6-hydroxymethyl-8-D-rib
pathogens and conversely are associated with inflammation. In contrast, the vitamin B9 metabolite acetyl-6-formylpterin binds as an antagonist to MR1, thus inhibiting
the activation of MAIT cells.

FIGURE 3 | Pivotal roles of vitamins B3, B7, and B9 in maintenance of immunological homeostasis. Vitamin B3 binds to GPR109a in dendritic cells and macrophages, and GPR109a signaling lead

improves via the promotion of keratinocyte proliferation and differentiation into fibroblasts (75). To maintain vitamin B5
Frontiers in Nutrition | www.frontiersin.org 6 April 2019 | Volume 6 | Article 48
Yoshii et al. Vitamin B Family and Host Immunity

levels during times of deficiency, CoA is converted back cysteamine inhibits peroxisome proliferator-activated receptor
to vitamin B5 or cysteamine via pantetheine (76). However, gamma (PPARγ) signaling, causing inflammation (77). Indeed,
colitis has been improved in pantetheine-producing-enzyme
knockout mice (78). Thus, vitamin B5 deficiency causes

Frontiers in Nutrition | www.frontiersin.org 7 April 2019 | Volume 6 | Article 48


Yoshii et al. Vitamin B Family and Host Immunity

inflammation through both dysfunction of the epithelial barrier arthritis (86). However, the mechanism underlying the regulation
and the production of pro-inflammatory molecules. of inflammation by vitamin B6 is currently unknown. Vitamin
In terms of immune responses, vitamin B5 enhances B6 contributes to intestinal immune regulation through the
protective activity against Mycobacterium tuberculosis infection metabolism of the lipid mediator sphingosine 1-phosphate (S1P).
by promoting innate immunity and adaptive immunity. In mice, S1P regulates lymphocyte trafficking into the intestines, especially
vitamin B5 supplementation activates phagocytosis and in the large intestine. Lymphocyte trafficking is dependent on S1P
cytokine production (including IL-6 and TNF-α) by macrophages gradient which is created by S1P production and its
and subsequently promotes Th1 and Th17 responses for the degradation. S1P degradation is mediated by S1P lyase that
clearance of M. tuberculosis from the lungs (79). Thus, vitamin requires vitamin B6 as a cofactor. The administration of vitamin
B5 contributes to host defense by activating immune B6 antagonist impairs S1P lyase activity and creates an
responses, suggesting that this vitamin has an important role inappropriate S1P gradient, resulted in impairing lymphocyte
as a natural adjuvant. migration from lymphoid tissues and reducing the numbers
Vitamin B5 is found in high concentrations as CoA or of lymphocytes in the intestines (87). The lymphocytes located
phosphopantetheine in liver, eggs, chicken, and fermented between gut epithelial cells are known as intraepithelial cells
soybeans. CoA and phosphopantetheine are converted to free (IELs) which are involved in the protection against pathogens
pantothenic acid by endogenous enzymes such as phosphatase (88). Therefore, vitamin B6 is important role for
and pantetheinase in the small intestine. Free pantothenic acid immunosurveillance in the intestines.
is absorbed via sodium-dependent multivitamin transporter Vitamin B6 is abundant in fish, chicken, tofu, sweet potato,
(SMVT) expressed on the epithelium of the small intestine and and avocado. Dietary vitamin B6 exists as PLP or PMP; it
is then released into the blood (80). Finally, free pantothenic acid is converted to free vitamin B6 by endogenous enzymes such
is converted back to CoA and distributed throughout the body, as pyridoxal phosphatase and is then absorbed by the small
particularly to the liver and kidney. intestine. Although absorption of vitamin B6 through acidic pH-
Bacterial vitamin B5 is synthesized from 2-dihydropantoate dependent and carrier-mediated transport has been shown, an
and β-alanine via de novo synthesis pathways (10). Bacterial intestinal pyridoxine transporter is yet to be identified in any
vitamin B5 exists as free pantothenic acid, which is directly mammalian species (11). After the absorption of free vitamin
absorbed in the large intestine, converted to CoA, and B6, it enters the blood and is converted back to PLP or PMP.
distributed in the same way as dietary vitamin B5. According Microbial vitamin B6 is synthesized as PLP from
to a genomic analysis, Bacteroides fragilis and Prevotella copri deoxyxylulose 5-phosphate and 4-phosphohydroxy-L-threonine
(Bacteroidetes); some Ruminococcus spp. (R. lactaris and R. or from glyceraldehyde-3-phosphate and D-ribulose 5-phosphate
torques) (Firmicutes); Salmonella enterica and Helicobacter pylori (10). In the large intestine, bacteria-derived PLP is converted
(Proteobacteria) possess a vitamin B5 biosynthesis pathway, to free vitamin B6, which is absorbed by passive transport,
indicating that intestinal commensal bacteria can produce transported to the blood, and distributed throughout the body.
vitamin B5. In contrast, most Fusobacterium (Fusobacteria) Metagenomic analysis has shown that Bacteroides fragilis
and Bifidobacterium spp. (Actinobacteria) and some strains of and Prevotella copri (Bacteroidetes), Bifidobacterium longum
Clostridium difficile, Faecalibacterium spp., and Lactobacillus spp. and, Collinsella aerofaciens (Actinobacteria), and Helicobacter
(Firmicutes) lack such a pathway (Table 1), although some of pylori (Proteobacteria) possess a vitamin B6 biosynthesis
them do express pantothenic acid transporter to utilize vitamin pathway. Bacteroidetes and Proteobacteria likely produce
B5 for energy generation (10), suggesting that these bacteria vitamin B6 starting from deoxyxylulose 5-phosphate and 4-
compete with the host for vitamin B5. phosphohydroxy-L-threonine, whereas Actinobacteria likely
start from glyceraldehyde-3-phosphate and D-ribulose 5-
phosphate. In contrast, most Firmicutes genera (Veillonella,
VITAMIN B6 Ruminococcus, Faecalibacterium, and Lactobacillus spp.),
except for some Clostridium (C. bartlettii, C. leptum, C.
Vitamin B6 exists in several forms, including as pyridoxine, methylpentosum, and C. sporogenes) and Lactobacillus spp. (L.
pyridoxal, and pyridoxamine. These forms of vitamin B6 are brevis and L. ruminis) lack a vitamin B6 biosynthesis pathway
precursors of the coenzymes pyridoxal phosphate (PLP) and (10) (Table 1).
pyridoxamine phosphate (PMP), which are involved in a variety
of cellular metabolic processes, including amino acid, lipid, and
carbohydrate metabolism (81). WHO/FAO recommends a daily VITAMIN B7
vitamin B6 intake of 1.3–1.7 mg for adults (16). Vitamin B6
deficiency is associated with the development of inflammatory Vitamin B7 (biotin) is a cofactor for several carboxylases that
diseases such as allergy and rheumatoid arthritis, as well as are essential for glucose, amino acid, and fatty acid metabolism
with neuronal dysfunction (82–84). Vitamin B6 deficiency (89). For example, vitamin B7 is an essential cofactor for acetyl-
disrupts the Th1–Th2 balance toward an excessive Th2 CoA carboxylase and fatty acid synthase, which are enzymes
response, resulting in allergy (85). Moreover, low plasma involved in fatty acid biosynthesis (90, 91). Thus, vitamin B7
vitamin B6 levels, together with increased levels of pro- likely influences immunometabolism. WHO/FAO recommends
inflammatory cytokines such as TNF-α and IL-6, have been a daily vitamin B7 intake of 30 µg for adults (16). Vitamin
observed in patients with rheumatoid

Frontiers in Nutrition | www.frontiersin.org 8 April 2019 | Volume 6 | Article 48


Yoshii et al. Vitamin B Family and Host Immunity

B7 suppresses gene expression by binding to (biotinylating) contributes to the maintenance of immunologic homeostasis.
histones; these genes include that encoding NF-κB, which is Regulatory T cells (Treg) express high levels of vitamin B9
a major signaling molecule for the production of several pro- receptor (folate receptor 4 [FR4]). Administration of anti-FR4
inflammatory cytokines (e.g., tumor necrosis factor alpha, IL- antibody results in specific reduction in the Treg cell population
1, IL-6, IL-8) (92, 93). Nuclear transcription of NF-κB is (106), suggesting that the vitamin B9–FR4 axis is required for
activated in response to vitamin B7 deficiency (94), suggesting Treg cell maintenance. In vitro culture of Treg cells under
that biotinylation of histones suppresses the expression of vitamin B9-reduced conditions leads to impaired cell survival,
genes encoding pro-inflammatory cytokines in NF-κB signaling with decreased expression of anti-apoptotic Bcl2 molecules,
(Figure 3). Therefore, vitamin B7 has anti-inflammatory effects although naïve T cells retain the ability to differentiate into
by inhibiting NF-κB activation, and dietary vitamin B7 Treg cells; this suggests that vitamin B9 is a survival factor for
deficiency causes inflammatory responses via enhanced Treg cells (87). Consistent with these findings, deficiency of
secretion of pro- inflammatory cytokines (95, 96). dietary vitamin B9 results in reduction of the Treg cell
Vitamin B7 is abundant in foods such as nuts, beans, and population in the small intestine (107, 108). Since Treg cells
oilseed. However, raw egg-white contains a large amount of play an important role in the prevention of excessive immune
avidin, which binds strongly to vitamin B7 and prevents its responses (109), mice fed a vitamin B9-deficient diet exhibit
absorption in the gut (97). Therefore, vitamin B7 deficiency can increased susceptibility to intestinal inflammation (107).
be caused not only by insufficient vitamin B7 intake, but also by Foods such as beef liver, green leafy vegetables, and asparagus
excessive intake of raw egg-white. Dietary biotin exists as a contain high levels of vitamin B9. Vitamin B9 exists as both
free protein-bound form or as biocytin (11). In the small mono- and polyglutamate folate species in the diet (110).
intestine, biotinidase releases free biotin from the bound Folate polyglutamate is deconjugated to the monoglutamate
protein and the free biotin is absorbed via the biotin form and then absorbed in the small intestine via folate
transporter SMVT (98). transporters such as proton-coupled folate transporter (PCFT)
Vitamin B7 is also produced by intestinal bacteria as (11, 111). In the intestinal epithelium, folate monoglutamate is
free biotin synthesized from malonyl CoA or pimelate via converted to tetrahydrofolate (THF), an active form and cofactor,
pimeloyl-CoA (99, 100). Bacterial free biotin is absorbed before being transported to the blood (111).
by SMVT expressed in the colon (23, 101). Metagenomic Intestinal bacteria synthesize vitamin B9 as THF from GTP,
analysis has shown that Bacteroides fragilis and Prevotella erythrose 4-phosphate, and phosphoenolpyruvate (38, 112).
copri (Bacteroidetes); Fusobacterium varium (Fusobacteria) Bacterial THF is directly absorbed in the colon via PCFT and
and Campylobacter coli (Proteobacteria) possess a vitamin distributed through the body by the blood (113). Metagenomic
B7 biosynthesis pathway (10). In contrast, Prevotella spp. analysis has shown that Bacteroides fragilis and Prevotella
(Bacteroidetes), Bifidobacterium spp. (Actinobacteria), and copri (Bacteroidetes); Clostridium difficile, Lactobacillus
Clostridium, Ruminococcus, Faecalibacterium, and Lactobacillus plantarum,
spp. (Firmicutes) lack such a pathway (Table 1); however, they L. reuteri, L. delbrueckii ssp. bulgaricus, and Streptococcus
do express free biotin transporter (10, 102), suggesting that thermophilus (Firmicutes), some species in Bifidobacterium spp
these bacteria also utilize dietary and bacterial vitamin B7 and (Actinobacteria); Fusobacterium varium (Fusobacteria) and
therefore may compete with the host. Thus, free biotin may Salmonella enterica (Proteobacteria) possess a folate biosynthesis
influence the composition of the gut microbiota, because biotin is pathway (Table 1) (10, 40). This suggests that almost all species
necessary for the growth and survival of the microbiota. in all phyla produce folate. Indeed, dietary supplementation
Indeed, biotin deficiency leads to gut dysbiosis and the with Bifidobacterium probiotic strains (B. adolescentis and
overgrowth of Lactobacillus murinus, leading to the B. pseudocatenulatum) enhances folate production in the
development of alopecia (103). Furthermore, vitamin B7 large intestine of folate-deficient rats and folate-free culture
production appears to proceed in a cooperative manner among medium (38, 41, 114). Furthermore, Lactobacillus plantarum,
different intestinal bacteria; Bifidobacterium longum in the L. delbrueckii ssp. bulgaricus, and L. reuteri enhance folate
intestine produces pimelate, which is a precursor of vitamin B7 production in bacterial culture supernatant lacking the
that enhances vitamin B7 production by other intestinal components needed for folate synthesis (38, 39, 115).
bacteria (104). In commensal bacteria, a vitamin B9 metabolite, 6-
formylpterin (6-FP), is produced by photodegradation
of folic acid (116). Like the vitamin B2 metabolite 6-
VITAMIN B9 hydroxymethyl-8-D-ribityllumazine, 6-FP binds to MR1,
but unlike 6-hydroxymethyl-8-D-ribityllumazine it cannot
Vitamin B9 (folate), in its active form as tetrahydrofolate, activate MAIT cells (62, 117). An analog of 6-FP, acetyl-6-FP,
is a cofactor in several metabolic reactions, including DNA is an antagonist of MR1, which inhibits MAIT cell activation
and amino acid synthesis. WHO/FAO recommends a daily (118). As mentioned in the section on vitamin B2, 6-
vitamin B9 intake of 400 µg for adults (16). Owing to the hydroxymethyl-8- D-ribityllumazine activates MAIT cells, which
high requirement of vitamin B9 by red blood cells, vitamin provide defense against pathogens, so vitamin B9 metabolites
B9 deficiency leads to megaloblastic anemia (23). Vitamin B9 may suppress excess MAIT cell responses and prevent
deficiency also inhibits the proliferation of human CD8 + T cells excessive allergic and inflammatory responses (Figure 2). The
in vitro by arresting the cell cycle in the S phase and increasing quantitative balance between dietary vitamin B2 and B9 and
the frequency of DNA damage (105). Moreover, vitamin B9 the composition of
Frontiers in Nutrition | www.frontiersin.org 9 April 2019 | Volume 6 | Article 48
Yoshii et al. Vitamin B Family and Host Immunity

the microbiota and its ability to metabolize these vitamins may (Table 1) (10, 32, 42, 43, 45). Indeed, Lactobacillus plantarum
be keys to understanding MAIT-cell-mediated homeostasis in and L. coryniformis isolated from fermented food produce
the intestine. vitamin B12 (33), and Bifidobacterium animalis synthesizes
FIGURE 4 | Schematic representation of B-vitamin-mediated interaction between commensal bacteria andduring
host immunity.
vitamin B12 milk fermentation (123).

VITAMIN B12
CONCLUSION
Vitamin B12 (cobalamin) is a cobalt-containing vitamin that,
in its active forms of methylcobalamin and adenosylcobalamin, B-vitamin-mediated immunological regulation is specific
catalyzes methionine synthesis (119). WHO/FAO recommends to different immune cells and immune responses: that is,
a daily vitamin B12 intake of 2.4 µg for adults (16). Together different B vitamins are required for different immune
with vitamin B6 and B9, vitamin B12 plays important roles responses (Figure 4). It was once thought that B vitamins were
in red blood cell formation and nucleic acid synthesis, obtained only from the diet; however, we know now that this is
especially in neurons. Therefore, vitamin B12 deficiency causes not the case and that the intestinal microbiota is also an
megaloblastic anemia and nervous system symptoms such important source of vitamins. Within the intestinal microbiota,
as numbness of the hands and feet (119). In terms of not all bacteria produce B vitamins and some bacteria utilize
host immunity, dietary vitamin B12 deficiency decreases the dietary B vitamins or B vitamins produced by other intestinal
number of CD8+ T cells and suppresses natural killer T- bacteria for their own needs; therefore, there may be
cell activity in mice; supplementation with methylcobalamin competition between the host and the intestinal microbiota for
improves these conditions (120), suggesting that vitamin B12 B vitamins (Figure 4). Research in this field is complicated,
contributes to the immune response via CD8+ T cells and because not only does the composition of the intestinal
natural killer T cells. microbiota vary among individuals, but also the composition of
Beef liver, bivalves, fish, chicken, and eggs contain high the diet can alter both the composition and function of the
levels of vitamin B12. Dietary vitamin B12 exists in complex intestinal microbiota. Therefore, vitamin-mediated
with dietary protein and is decomposed to free vitamin B12 by immunological maintenance also varies among individuals.
pepsin in the stomach. Free vitamin B12 is absorbed by the Further examinations in this field are needed, and the new
epithelial cells of the small intestine via intrinsic factor (IF), a information uncovered will help to develop a new era of
gastric glycoprotein. Inside the epithelial cells, IF-vitamin B12 precision health and nutrition.
complex is decomposed to free vitamin B12 by lysosome and then
released into the blood, where it is converted to the active AUTHOR CONTRIBUTIONS
form and distributed throughout the body (121, 122).
Bacterial vitamin B12 is synthesized from precorrin- KY and KH wrote the draft of review article which was
2 to produce adenosylcobalamin (10), which is absorbed corrected by JK. KY, KH, and KS drew figures and JK performed
directly by the large intestine and distributed throughout the correction.
body; the mechanism underlying this absorption is currently
unclear. Metagenomic analysis has predicted that Bacteroides
ACKNOWLEDGMENTS
fragilis and Prevotella copri (Bacteroidetes); Clostridium
difficile, Faecalibacterium prausnitzii and Ruminococcus This review article contains results obtained from our studies
lactaris (Firmicutes); Bifidobacterium animalis, B.infantis, that were supported at least in part by grants from the
and B.longum (Actinobacteria); Fusobacterium varium Japan Agency for Medical Research and Development
(Fusobacteria) possess a vitamin B12 biosynthesis pathway [AMED; 17fk0108223h0002 (JK), 17ek0410032s0102
(JK), 17fk0108207h0002 (JK), 17ek0210078h0002 (JK),

Frontiers in Nutrition | www.frontiersin.org 1 April 2019 | Volume 6 | Article 48


0
Yoshii et al. Vitamin B Family and Host Immunity

17ak0101068h0001 (JK), 17gm1010006s0101 (JK), and Promotion of Science [JSPS, JP16H01373 (JK), JP15H05790
19ek0410062h0001 (JK)]; Cross-ministerial Strategic Innovation (JK), JP17H04134 (JK), JP26670241 (JK), JP26293111 (JK),
Promotion Program; the Ministry of Health, Labor, and JP18K17997 (KH), and JP18J00556 (KH)]; the ONO Medical
Welfare of Japan; the Science and Technology Research Research Foundation (JK); the Canon Foundation (JK); the
Promotion Program for Agriculture, Forestry, Fisheries, and Terumo Foundation for the Life Sciences and Arts; and the
Food Industry; the Ministry of Education, Culture, Sports, Nippon Ham Foundation for the Future of Food. KH is a JSPS
Science, and Technology of Japan; the Japan Society for the Research Fellow.

REFERENCES 20. Shikina T, Hiroi T, Iwatani K, Jang MH, Fukuyama S, Tamura M, et al.
IgA class switch occurs in the organized nasopharynx- and gut-associated
1. Belkaid Y, Harrison OJ. Homeostatic immunity and the microbiota. lymphoid tissue, but not in the diffuse lamina propria of airways and gut. J
Immunity. (2017) 46:562–76. doi: 10.1016/j.immuni.2017.04.008 Immunol. (2004) 172:6259–64. doi: 10.4049/jimmunol.172.10.6259
2. Hirata S-I, Kunisawa J. Gut microbiome, metabolome, and allergic 21. Kunisawa J, Sugiura Y, Wake T, Nagatake T, Suzuki H, Nagasawa R, et al.
diseases. Allergol Int Off J Jpn Soc Allergol. (2017) 66:523–8. Mode of bioenergetic metabolism during B cell differentiation in the intestine
doi: 10.1016/j.alit.2017.06.008 determines the distinct requirement for vitamin B1. Cell Rep. (2015) 13:122–
3. Shibata N, Kunisawa J, Kiyono H. Dietary and microbial metabolites 31. doi: 10.1016/j.celrep.2015.08.063
in the regulation of host immunity. Front Microbiol. (2017) 8:2171. 22. Rindi G, Laforenza U. Thiamine intestinal transport and related issues:
doi: 10.3389/fmicb.2017.02171 recent aspects. Proc Soc Exp Biol Med Soc Exp Biol Med. (2000) 224:246–
4. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, 55. doi: 10.1046/j.1525-1373.2000.22428.x
the gut microbiome, and immune system: envisioning the future. Nature. 23. Said HM. Recent advances in transport of water-soluble vitamins
(2011) 474:327–36. doi: 10.1038/nature10213 in organs of the digestive system: a focus on the colon and the
5. Lamichhane A, Kiyono H, Kunisawa J. Nutritional components regulate pancreas. Am J Physiol Gastrointest Liver Physiol. (2013) 305:G601–10.
the gut immune system and its association with intestinal immune doi: 10.1152/ajpgi.00231.2013
disease development. J Gastroenterol Hepatol. (2013) 28(Suppl. 4):18–24. 24. Barrow BJ, Ortiz-Reyes R, O’Riordan MA, Pretlow TP. In situ localization of
doi: 10.1111/jgh.12259 enzymes and mucin in normal rat colon embedded in plastic. Histochem J.
6. Whatham A, Bartlett H, Eperjesi F, Blumenthal C, Allen J, Suttle C, (1989) 21:289–95. doi: 10.1007/BF01757182
et al. Vitamin and mineral deficiencies in the developed world and their 25. Nabokina SM, Inoue K, Subramanian VS, Valle JE, Yuasa H, Said HM.
effect on the eye and vision. Ophthalmic Physiol Opt. (2008) 28:1–12. Molecular identification and functional characterization of the human
doi: 10.1111/j.1475-1313.2007.00531.x colonic thiamine pyrophosphate transporter. J Biol Chem. (2014)
7. Lee W-J, Hase K. Gut microbiota-generated metabolites in animal health and 289:4405– 16. doi: 10.1074/jbc.M113.528257
disease. Nat Chem Biol. (2014) 10:416–24. doi: 10.1038/nchembio.1535 26. Lindhurst MJ, Fiermonte G, Song S, Struys E, De Leonardis F,
8. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Schwartzberg PL, et al. Knockout of Slc25a19 causes mitochondrial
Nat Rev Immunol. (2016) 16:341–52. doi: 10.1038/nri.2016.42 thiamine pyrophosphate depletion, embryonic lethality, CNS
9. Shi N, Li N, Duan X, Niu H. Interaction between the gut malformations, and anemia. Proc Natl Acad Sci USA. (2006) 103:15927–32.
microbiome and mucosal immune system. Mil Med Res. (2017) 4:14. doi: 10.1073/pnas.0607661103
doi: 10.1186/s40779-017-0122-9 27. Jurgenson CT, Begley TP, Ealick SE. The structural and biochemical
10. Magnúsdóttir S, Ravcheev D, de Crécy-Lagard V, Thiele I. Systematic foundations of thiamin biosynthesis. Annu Rev Biochem. (2009) 78:569–
genome assessment of B-vitamin biosynthesis suggests co-operation 603. doi: 10.1146/annurev.biochem.78.072407.102340
among gut microbes. Front Genet. (2015) 6:148. doi: 28. Leonardi R, Roach PL. Thiamine Biosynthesis in Escherichia coli in
10.3389/fgene.2015.00148 vitro reconstruction of the thiazole synthase activity. J Biol Chem. (2004)
11. Said HM. Intestinal absorption of water-soluble vitamins in health and 279:17054–62. doi: 10.1074/jbc.M312714200
disease. Biochem J. (2011) 437:357–72. doi: 10.1042/BJ20110326 29. McRose D, Guo J, Monier A, Sudek S, Wilken S, Yan S, et al. Alternatives to
12. Hosomi K, Kunisawa J. The specific roles of vitamins in the regulation of vitamin B1 uptake revealed with discovery of riboswitches in multiple marine
immunosurveillance and maintenance of immunologic homeostasis in the eukaryotic lineages. ISME J. (2014) 8:2517–29. doi: 10.1038/ismej.2014.146
gut. Immune Netw. (2017) 17:13–9. doi: 10.4110/in.2017.17.1.13 30. Taylor SV, Kelleher NL, Kinsland C, Chiu HJ, Costello CA, Backstrom
13. Suzuki H, Kunisawa J. Vitamin-mediated immune regulation in the AD, et al. Thiamin biosynthesis in Escherichia coli. Identification of
development of inflammatory diseases. Endocr Metab Immune Disord ThiS thiocarboxylate as the immediate sulfur donor in the thiazole
Drug Targets. (2015) 15:212–5. doi: 10.2174/1871530315666150316122128 formation J Biol Chem. (1998) 273:16555–60. doi: 10.1074/jbc.273.
14. Frank RAW, Leeper FJ, Luisi BF. Structure, mechanism and catalytic duality 26.16555
of thiamine-dependent enzymes. Cell Mol Life Sci. (2007) 64:892–905. 31. Drywien M, Frackiewicz J, Górnicka M, Gadek J, Jałosinska M. Effect of
doi: 10.1007/s00018-007-6423-5 probiotic and storage time of thiamine and riboflavin content in the milk
15. Huskisson E, Maggini S, Ruf M. The role of vitamins and minerals drinks fermented by Lactobacillus casei KNE-1. Rocz Panstw Zakl Hig.
in energy metabolism and well-being. J Int Med Res. (2007) 35:277–89. (2015) 66:373–7.
doi: 10.1177/147323000703500301 32. Deguchi Y, Morishita T, Mutai M. Comparative studies on synthesis of
16. WHO/FAO. Vitamin and Mineral Requirements in Human Nutrition. 2nd water- soluble vitamins among human species of Bifidobacteria. Agric Biol
ed. Report of a joint WHO/FAO Expert Consultation, Bangkok, Thailand Chem. (1985) 49:13–9.
1998. Geneva: World Health Organization/Rome: Food and Agriculture 33. Masuda M, Ide M, Utsumi H, Niiro T, Shimamura Y, Murata M. Production
Organization of the United Nations (2004). potency of folate, vitamin B12, and thiamine by lactic acid bacteria
17. Mathis D, Shoelson SE. Immunometabolism: an emerging frontier. Nat Rev isolated from Japanese pickles. Biosci Biotechnol Biochem. (2012) 76:2061–7.
Immunol. (2011) 11:81–3. doi: 10.1038/nri2922 doi: 10.1271/bbb.120414
18. Buck MD, Sowell RT, Kaech SM, Pearce EL. Metabolic Instruction of 34. Levit R, Savoy de Giori G, de Moreno de LeBlanc A, LeBlanc JG. Evaluation
Immunity. Cell. (2017) 169:570–86. doi: 10.1016/j.cell.2017.04.004 of the effect of soymilk fermented by a riboflavin-producing Lactobacillus
19. Pearce EL, Poffenberger MC, Chang C-H, Jones RG. Fueling immunity: plantarum strain in a murine model of colitis. Benef Microbes. (2016)
insights into metabolism and lymphocyte function. Science. (2013) 8:65– 72. doi: 10.3920/BM2016.0063
342:1242454. doi: 10.1126/science.1242454

Frontiers in Nutrition | www.frontiersin.org 1 April 2019 | Volume 6 | Article 48


1
Yoshii et al. Vitamin B Family and Host Immunity

35. Juarez Del Valle M, Lai-o JE, de Moreno de LeBlanc A, Savoy de 56. Duurkens RH, Tol MB, Geertsma ER, Permentier HP, Slotboom DJ. Flavin
Giori G, LeBlanc JG. Soyamilk fermented with riboflavin-producing binding to the high affinity riboflavin transporter RibU. J Biol Chem. (2007)
Lactobacillus plantarum CRL 2130 reverts and prevents ariboflavinosis in 282:10380–6. doi: 10.1074/jbc.M608583200
murine models. Br J Nutr. (2016) 116:1229–35. doi: 57. Chen Z, Wang H, D’Souza C, Sun S, Kostenko L, Eckle SBG, et al. Mucosal-
10.1017/S00071145160 03378 associated invariant T-cell activation and accumulation after in vivo
36. Russo P, Capozzi V, Arena MP, Spadaccino G, Due-as MT, López P, infection depends on microbial riboflavin synthesis and co-stimulatory
et al. Riboflavin-overproducing strains of Lactobacillus fermentum for signals. Mucosal Immunol. (2017) 10:58–68. doi: 10.1038/mi.2016.39
riboflavin-enriched bread. Appl Microbiol Biotechnol. (2014) 98:3691–700. 58. Gold MC, Cerri S, Smyk-Pearson S, Cansler ME, Vogt TM, Delepine J, et al.
doi: 10.1007/s00253-013-5484-7 Human mucosal associated invariant T cells detect bacterially infected
37. Shah N, Patel A. Recent advances in biosynthesis of vitamin and enzyme cells. PLoS Biol. (2010) 8:e1000407. doi: 10.1371/journal.pbio.1000407
from food grade bacteria. Intl J Food Ferment Technol. (2014) 4:79–85. 59. Ussher JE, Klenerman P, Willberg CB. Mucosal-associated invariant T-
doi: 10.5958/2321-7111.2015.00001.3 cells: new players in anti-bacterial immunity. Front Immunol. (2014) 5:450.
38. Rossi M, Amaretti A, Raimondi S. Folate production by probiotic bacteria. doi: 10.3389/fimmu.2014.00450
Nutrients. (2011) 3:118–34. doi: 10.3390/nu3010118 60. Le Bourhis L, Dusseaux M, Bohineust A, Bessoles S, Martin E, Premel V,
39. Homayouni Rad A, Yari Khosroushahi A, Khalili M, Jafarzadeh S. Folate bio- et al. MAIT cells detect and efficiently lyse bacterially-infected epithelial cells.
fortification of yoghurt and fermented milk: a review. Dairy Sci Technol. PLoS Pathog. (2013) 9:e1003681. doi: 10.1371/journal.ppat.1003681
(2016) 96:427–41. doi: 10.1007/s13594-016-0286-1 61. Thakur K, Tomar SK, De S. Lactic acid bacteria as a cell factory
40. Crittenden RG, Martinez NR, Playne MJ. Synthesis and utilisation of folate for riboflavin production. Microb Biotechnol. (2015) 9:441–51.
by yoghurt starter cultures and probiotic bacteria. Int J Food Microbiol. doi: 10.1111/1751-7915.12335
(2003) 80:217–22. doi: 10.1016/S0168-1605(02)00170-8 62. Kjer-Nielsen L, Patel O, Corbett AJ, Le Nours J, Meehan B, Liu L, et al.
41. D’Aimmo MR, Mattarelli P, Biavati B, Carlsson NG, Andlid T. The potential MR1 presents microbial vitamin B metabolites to MAIT cells. Nature.
of bifidobacteria as a source of natural folate. J Appl Microbiol. (2012) (2012) 491:717–23. doi: 10.1038/nature11605
112:975–84. doi: 10.1111/j.1365-2672.2012.05261.x 63. Serriari N-E, Eoche M, Lamotte L, Lion J, Fumery M, Marcelo P, et al. Innate
42. Song H, Yoo Y, Hwang J, Na Y-C, Kim HS. Faecalibacterium mucosal-associated invariant T (MAIT) cells are activated in inflammatory
prausnitzii subspecies-level dysbiosis in the human gut microbiome bowel diseases. Clin Exp Immunol. (2014) 176:266–74. doi: 10.1111/cei.12277
underlying atopic dermatitis. J Allergy Clin Immunol. (2016) 137:852–60. 64. Chai JT, Digby JE, Choudhury RP. GPR109A and vascular inflammation.
doi: 10.1016/j.jaci.2015.08.021 Curr Atheroscler Rep. (2013) 15:9. doi: 10.1007/s11883-013-0325-9
43. Piwowarek K, Lipinska E, Hac´-Szymanczuk E, Kieliszek M, Scibisz 65. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, et al. Activation
I. Propionibacterium spp.-source of propionic acid, vitamin B12, and other of Gpr109a, receptor for niacin and the commensal metabolite butyrate,
metabolites important for the industry. Appl Microbiol Biotechnol. (2018) suppresses colonic inflammation and carcinogenesis. Immunity. (2014)
102:515–38. doi: 10.1007/s00253-017-8616-7 40:128–39. doi: 10.1016/j.immuni.2013.12.007
44. Gu Q, Zhang C, Song D, Li P, Zhu X. Enhancing vitamin B12 content in soy- 66. Lipszyc PS, Cremaschi GA, Zubilete MZ, Bertolino MLA, Capani F,
yogurt by Lactobacillus reuteri. Int J Food Microbiol. (2015) 206:56–9. doi: Genaro AM, et al. Niacin modulates pro-inflammatory cytokine secretion.
10.1016/j.ijfoodmicro.2015.04.033 A potential mechanism involved in its anti-atherosclerotic effect Open
45. Lee J-H, O’Sullivan DJ. Genomic insights into bifidobacteria. Microbiol Mol Cardiovasc Med J. (2013) 7:90–8. doi: 10.2174/1874192401307010090
Biol Rev MMBR. (2010) 74:378–416. doi: 10.1128/MMBR.00004-10 67. Amanullah S, Seeber C. Niacin deficiency resulting in neuropsychiatric
46. Costliow ZA, Degnan PH. Thiamine acquisition strategies impact symptoms: A case study and review of literature. Clin Neuropsychiatry.
metabolism and competition in the gut microbe Bacteroides (2010) 7:10–14.
thetaiotaomicron. mSystems. (2017) 2:17. doi: 10.1128/mSystems.00116-17 68. Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, et al. Impact of the gut microbiota
47. Almeida L, Lochner M, Berod L, Sparwasser T. Metabolic pathways in T on intestinal immunity mediated by tryptophan metabolism. Front Cell Infect
cell activation and lineage differentiation. Semin Immunol. (2016) 28:514–24. Microbiol. (2018) 8:13. doi: 10.3389/fcimb.2018.00013
doi: 10.1016/j.smim.2016.10.009 69. Gazzaniga F, Stebbins R, Chang SZ, McPeek MA, Brenner C. Microbial NAD
48. Schramm M, Wiegmann K, Schramm S, Gluschko A, Herb M, Utermöhlen metabolism: lessons from comparative genomics. Microbiol Mol Biol Rev.
O, et al. Riboflavin (vitamin B2) deficiency impairs NADPH oxidase 2 (Nox2) (2009) 73:529–41. doi: 10.1128/MMBR.00042-08
priming and defense against Listeria monocytogenes. Eur J Immunol. 70. Kurnasov O, Goral V, Colabroy K, Gerdes S, Anantha S, Osterman
(2014) 44:728–41. doi: 10.1002/eji.201343940 A, et al. NAD biosynthesis: identification of the tryptophan to
49. Pinto JT, Zempleni J. Riboflavin. Adv Nutr. (2016) 7:973–5. quinolinate pathway in bacteria. Chem Biol. (2003) 10:1195–204.
doi: 10.3945/an.116.012716 doi: 10.1016/j.chembiol.2003.11.011
50. Sundaram U. Regulation of intestinal vitamin B2 absorption. 71. Rodionov DA, Li X, Rodionova IA, Yang C, Sorci L, Dervyn E, et al.
Focus on “Riboflavin uptake by human-derived colonic epithelial Transcriptional regulation of NAD metabolism in bacteria: genomic
NCM460 cells.” Am J Physiol Cell Physiol. (2000) 278:C268–9. reconstruction of NiaR (YrxA) regulon. Nucleic Acids Res. (2008) 36:2032–
doi: 10.1152/ajpcell.2000.278.2.C268 46. doi: 10.1093/nar/gkn046
51. Jaeger B, Bosch AM. Clinical presentation and outcome of riboflavin 72. Kennedy DO. B Vitamins and the brain: mechanisms, dose and efficacy—a
transporter deficiency: mini review after five years of experience. J Inherit review. Nutrients. (2016) 8:68. doi: 10.3390/nu8020068
Metab Dis. (2016) 39:559–64. doi: 10.1007/s10545-016-9924-2 73. Hodges RE, Ohlson MA, Bean WB. Pantothenic acid deficiency in man1. J
52. Subramanian VS, Lambrecht N, Lytle C, Said HM. Conditional (intestinal- Clin Invest. (1958) 37:1642–57. doi: 10.1172/JCI103756
specific) knockout of the riboflavin transporter-3 (RFVT-3) impairs 74. Yang M, Moclair B, Hatcher V, Kaminetsky J, Mekas M, Chapas A, et al. A
riboflavin absorption. Am J Physiol - Gastrointest Liver Physiol. (2016) randomized, double-blind, placebo-controlled study of a novel
310:G285–93. doi: 10.1152/ajpgi.00340.2015 pantothenic Acid-based dietary supplement in subjects with mild to moderate
53. Yonezawa A, Masuda S, Katsura T, Inui K. Identification and functional facial acne. Dermatol Ther. (2014) 4:93–101. doi: 10.1007/s13555-014-
characterization of a novel human and rat riboflavin transporter, RFT1. Am 0052-3
J Physiol Cell Physiol. (2008) 295:C632–641. doi: 10.1152/ajpcell.00019.2008 75. Kobayashi D, Kusama M, Onda M, Nakahata N. The effect of pantothenic
54. García-Angulo VA. Overlapping riboflavin supply pathways in bacteria. Crit acid deficiency on keratinocyte proliferation and the synthesis of
Rev Microbiol. (2017) 43:196–209. doi: 10.1080/1040841X.2016.1192578 keratinocyte growth factor and collagen in fibroblasts. J Pharmacol Sci.
55. Tastan C, Karhan E, Zhou W, Fleming E, Voigt AY, Yao X, et al. Tuning (2011) 115:230–4. doi: 10.1254/jphs.10224SC
of human MAIT cell activation by commensal bacteria species and MR1- 76. Leonardi R, Zhang Y-M, Rock CO, Jackowski S. Coenzyme A: back in action.
dependent T-cell presentation. Mucosal Immunol. (2018) 11:1591–605. Prog Lipid Res. (2005) 44:125–53. doi: 10.1016/j.plipres.2005.04.001
doi: 10.1038/s41385-018-0072-x 77. Zhang B, Lo C, Shen L, Sood R, Jones C, Cusmano-Ozog K,
et al. The role of vanin-1 and oxidative stress–related pathways in

Frontiers in Nutrition | www.frontiersin.org 1 April 2019 | Volume 6 | Article 48


2
Yoshii et al. Vitamin B Family and Host Immunity

distinguishing acute and chronic pediatric ITP. Blood. (2011) 117:4569–79. 99. Bi H, Zhu L, Jia J, Cronan JE. A Biotin biosynthesis gene restricted to
doi: 10.1182/blood-2010-09-304931 Helicobacter. Sci Rep. (2016) 6:21162. doi: 10.1038/srep21162
78. Berruyer C, Pouyet L, Millet V, Martin FM, LeGoffic A, Canonici 100. Entcheva P, Phillips DA, Streit WR. Functional analysis of Sinorhizobium
A, et al. Vanin-1 licenses inflammatory mediator production by gut meliloti genes involved in biotin synthesis and transport. Appl Environ
epithelial cells and controls colitis by antagonizing peroxisome Microbiol. (2002) 68:2843–8. doi: 10.1128/AEM.68.6.2843-2848.2002
proliferator- activated receptor gamma activity. J Exp Med. (2006) 101. Sabui S, Bohl JA, Kapadia R, Cogburn K, Ghosal A, Lambrecht NW, et al.
203:2817–27. doi: 10.1084/jem.20061640 Role of the sodium-dependent multivitamin transporter (SMVT) in the
79. He W, Hu S, Du X, Wen Q, Zhong X-P, Zhou X, et al. Vitamin B5 reduces maintenance of intestinal mucosal integrity. Am J Physiol Gastrointest
bacterial growth via regulating innate immunity and adaptive immunity Liver Physiol. (2016) 311:G561–70. doi: 10.1152/ajpgi.00240.2016
in mice infected with Mycobacterium tuberculosis. Front Immunol. (2018) 102. Fisher DJ, Fernández RE, Adams NE, Maurelli AT. Uptake of biotin
9:365. doi: 10.3389/fimmu.2018.00365 by Chlamydia Spp. through the use of a bacterial transporter (BioY)
80. Quick M, Shi L. The sodium/multivitamin transporter: a multipotent and a host-cell transporter (SMVT). PLoS ONE. (2012) 7:e46052.
system with therapeutic implications. Vitam Horm. (2015) 98:63–100. doi: 10.1371/journal.pone.0046052
doi: 10.1016/bs.vh.2014.12.003 103. Hayashi A, Mikami Y, Miyamoto K, Kamada N, Sato T, Mizuno S,
81. Parra M, Stahl S, Hellmann H. Vitamin B6 and its role in cell metabolism et al. Intestinal dysbiosis and biotin deprivation induce alopecia through
and physiology. Cells. (2018) 7:84. doi: 10.3390/cells7070084 overgrowth of Lactobacillus murinus in mice. Cell Rep. (2017) 20:1513–24.
82. Doke S, Inagaki N, Hayakawa T, Tsuge H. Effect of vitamin B6 deficiency on doi: 10.1016/j.celrep.2017.07.057
an antibody production in mice. Biosci Biotechnol Biochem. (1997) 104. Sugahara H, Odamaki T, Fukuda S, Kato T, Xiao J, Abe F, et al.
61:1331– 6. doi: 10.1271/bbb.61.1331 Probiotic Bifidobacterium longum alters gut luminal metabolism through
83. Huang S-C, Wei JC-C, Wu DJ, Huang Y-C. Vitamin B (6) supplementation modification of the gut microbial community. Sci Rep. (2015) 5:13548.
improves pro-inflammatory responses in patients with rheumatoid doi: 10.1038/srep13548
arthritis. Eur J Clin Nutr. (2010) 64:1007–13. doi: 10.1038/ejcn.2010.107 105. Courtemanche C, Elson-Schwab I, Mashiyama ST, Kerry N, Ames BN.
84. Jannusch K, Jockwitz C, Bidmon H-J, Moebus S, Amunts K, Caspers S. A Folate deficiency inhibits the proliferation of primary human CD8+
complex interplay of vitamin B1 and B6 metabolism with cognition, brain
T lymphocytes in vitro. J Immunol Baltim Md. (2004) 173:3186–92.
structure, and functional connectivity in older adults. Front Neurosci. doi: 10.4049/jimmunol.173.5.3186
(2017) 11:596. doi: 10.3389/fnins.2017.00596 106. Yamaguchi T, Hirota K, Nagahama K, Ohkawa K, Takahashi T, Nomura
85. Qian B, Shen S, Zhang J, Jing P. Effects of vitamin B6 deficiency on the T, et al. Control of immune responses by antigen-specific regulatory
composition and functional potential of T cell populations. J Immunol Res. T cells expressing the folate receptor. Immunity. (2007) 27:145–59.
(2017) 2017:2197975. doi: 10.1155/2017/2197975 doi: 10.1016/j.immuni.2007.04.017
86. Chiang E-PI, Selhub J, Bagley PJ, Dallal G, Roubenoff R. Pyridoxine 107. Kinoshita M, Kayama H, Kusu T, Yamaguchi T, Kunisawa J, Kiyono H, et al.
supplementation corrects vitamin B6 deficiency but does not improve Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the
inflammation in patients with rheumatoid arthritis. Arthritis Res Ther. colon. J Immunol. (2012) 189:2869–78. doi: 10.4049/jimmunol.1200420
(2005) 7:R1404–11. doi: 10.1186/ar1839 108. Kunisawa J, Hashimoto E, Ishikawa I, Kiyono H. A pivotal role of vitamin
87. Kunisawa J, Kiyono H. Vitamin-mediated regulation of intestinal immunity. B9 in the maintenance of regulatory T cells in vitro and in vivo. PLoS ONE.
Front Immunol. (2013) 4:189. doi: 10.3389/fimmu.2013.00189 (2012) 7:e32094. doi: 10.1371/journal.pone.0032094
88. Sheridan BS, Lefrançois L. Intraepithelial lymphocytes: To serve and protect. 109. Sakaguchi S, Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T. Regulatory
Curr Gastroenterol Rep. (2010) 12:513–21. doi: 10.1007/s11894-010-0148-6 T cells: how do they suppress immune responses? Int Immunol. (2009)
89. Tong L. Structure and function of biotin-dependent carboxylases. Cell Mol 21:1105–11. doi: 10.1093/intimm/dxp095
Life Sci. (2013) 70:863–91. doi: 10.1007/s00018-012-1096-0 110. Melse-Boonstra A, de Bree A, Verhoef P, Bjørke-Monsen AL, Verschuren
90. Ikeda M, Miyamoto A, Mutoh S, Kitano Y, Tajima M, Shirakura WMM. Dietary monoglutamate and polyglutamate folate are associated with
D, et al. Development of biotin prototrophic and hyper-auxotrophic plasma folate concentrations in Dutch men and women aged 20-65 years.
Corynebacterium glutamicum strains toward biotin production. Appl Env J Nutr. (2002) 132:1307–12. doi: 10.1093/jn/132.6.1307
Microbiol. (2013) 2013:828. doi: 10.1128/AEM.00828-13 111. Zhao R, Diop-Bove N, Visentin M, Goldman ID. Mechanisms of membrane
91. Ikeda M, Nagashima T, Nakamura E, Kato R, Ohshita M, Hayashi M, et al. transport of folates into cells and across epithelia. Annu Rev Nutr. (2011)
In vivo roles of fatty acid biosynthesis enzymes in biosynthesis of biotin 31:177–201. doi: 10.1146/annurev-nutr-072610-145133
and α- lipoic acid in Corynebacterium glutamicum. Appl Environ Microbiol. 112. Maguire F, Henriquez F L, Leonard G, Dacks JB, Brown MW, Richards
(2017) 83:17. doi: 10.1128/AEM.01322-17 TA. Complex patterns of gene fission in the eukaryotic folate biosynthesis
92. Kothapalli N, Camporeale G, Kueh A, Chew YC, Oommen AM, Griffin JB, pathway. Genome Biol Evol. (2014) 6:2709–20. doi: 10.1093/gbe/evu213
et al. Biological functions of biotinylated histones. J Nutr Biochem. (2005) 113. Maynard C, Cummins I, Green J, Weinkove D. A bacterial route for folic
16:446–8. doi: 10.1016/j.jnutbio.2005.03.025 acid supplementation. BMC Biol. (2018) 16:3. doi: 10.1186/s12915-018-
93. Liu T, Zhang L, Joo D, Sun S-C. NF-κB signaling in inflammation. Signal 0534-3
Transduct Target Ther. (2017) 2:17023. doi: 10.1038/sigtrans.2017.23 114. Strozzi GP, Mogna L. Quantification of folic acid in human feces after
94. Rodriguez-Melendez R, Zempleni J. Regulation of gene administration of Bifidobacterium probiotic strains. J Clin Gastroenterol.
expression by biotin (review). J Nutr Biochem. (2003) 14:680–90. (2008) 42(Suppl. 3):S179–84. doi: 10.1097/MCG.0b013e31818087d8
doi: 10.1016/j.jnutbio.2003.07.001 115. Santos F, Wegkamp A, de Vos WM, Smid EJ, Hugenholtz J. High-
95. Agrawal S, Agrawal A, Said HM. Biotin deficiency enhances the level folate production in fermented foods by the B12 producer
inflammatory response of human dendritic cells. Am J Physiol Cell Physiol. Lactobacillus reuteri JCM1112. Appl Environ Microbiol. (2008) 74:3291–4.
(2016) 311:C386–391. doi: 10.1152/ajpcell.00141.2016 doi: 10.1128/AEM.02719-07
96. Elahi A, Sabui S, Narasappa NN, Agrawal S, Lambrecht NW, Agrawal A, 116. Araújo MM, Marchioni E, Villavicencio ALCH, Zhao M, di Pascoli T, Kuntz
et al. Biotin deficiency induces Th1- and Th17-mediated proinflammatory
F, et al. Mechanism of folic acid radiolysis in aqueous solution. LWT - Food
responses in human CD4+ T lymphocytes via activation of the Sci Technol. (2015) 63:599–603. doi: 10.1016/j.lwt.2015.03.038
mTOR signaling pathway. J Immunol Baltim Md. (2018) 200:2563–70. 117. Patel O, Kjer-Nielsen L, Le Nours J, Eckle SBG, Birkinshaw R, Beddoe T,
doi: 10.4049/jimmunol.1701200 et al. Recognition of vitamin B metabolites by mucosal-associated
97. Zempleni J, Hassan YI, Wijeratne SS. Biotin and biotinidase invariant T cells. Nat Commun. (2013) 4:2142. doi: 10.1038/ncomms3142
deficiency. Expert Rev Endocrinol Metab . (2008) 3:715–24. 118. Eckle SBG, Birkinshaw RW, Kostenko L, Corbett AJ, McWilliam
doi: 10.1586/17446651.3.6.715 HEG, Reantragoon R, et al. A molecular basis underpinning
98. Zempleni J, Wijeratne SSK, Hassan YI. Biotin. BioFactors Oxf Engl. (2009) the T cell receptor heterogeneity of mucosal-associated invariant
35:36–46. doi: 10.1002/biof.8 T cells. J Exp Med. (2014) 211:1585–600. doi: 10.1084/jem.
20140484

Frontiers in Nutrition | www.frontiersin.org 1 April 2019 | Volume 6 | Article 48


3
Yoshii et al. Vitamin B Family and Host Immunity

119. Bito T, Watanabe F. Biochemistry, function, and deficiency of vitamin 123. Patel A, Shah N, Prajapati JB. Biosynthesis of vitamins and enzymes in
B12 in Caenorhabditis elegans. Exp Biol Med. (2016) 241:1663–8. fermented foods by lactic acid bacteria and related genera - a promising
doi: 10.1177/1535370216662713 approach. Croat J Food Sci Technol. (2013) 5:85–91.
120. Tamura J, Kubota K, Murakami H, Sawamura M, Matsushima T,
Tamura T, et al. Immunomodulation by vitamin B12: augmentation
Conflict of Interest Statement: KS was employed by Nippon Flour Mills Co., Ltd.
of CD8+ T lymphocytes and natural killer (NK) cell activity
in vitamin B12-deficient patients by methyl-B12 treatment. Clin The remaining authors declare that the research was conducted in the absence
Exp Immunol. (1999) 116:28–32. doi: 10.1046/j.1365-2249.1999. of any commercial or financial relationships that could be construed as a
00870.x potential conflict of interest.
121. Beedholm-Ebsen R, van de Wetering K, Hardlei T, Nexø E, Borst
P, Moestrup SK. Identification of multidrug resistance protein Copyright © 2019 Yoshii, Hosomi, Sawane and Kunisawa. This is an open-access
1 (MRP1/ABCC1) as a molecular gate for cellular export of article distributed under the terms of the Creative Commons Attribution License (CC
cobalamin. Blood. (2010) 115:1632–9. doi: 10.1182/blood-2009-07- BY). The use, distribution or reproduction in other forums is permitted, provided
232587 the original author(s) and the copyright owner(s) are credited and that the
122. Green R. Vitamin B12 deficiency from the perspective original publication in this journal is cited, in accordance with accepted academic
of a practicing hematologist. Blood. (2017) 129:2603–11. practice. No use, distribution or reproduction is permitted which does not comply
doi: 10.1182/blood-2016-10-569186 with these terms.

Frontiers in Nutrition | www.frontiersin.org 1 April 2019 | Volume 6 | Article 48


4

You might also like