ArticlePDF Available

Zinc Supplementation Increases Zinc Status and Thymopoiesis in Aged Mice

Authors:

Abstract and Figures

The age-related decline in lymphocyte development and function coincides with impaired zinc status in the elderly. Thymic involution and reduced immune responsiveness are classic hallmarks of both aging and zinc deficiency, resulting in decreased host defense and an increased susceptibility to infections. Thus, compromised zinc status associated with aging may be an important contributing factor in reduced thymopoiesis and impaired immune functions. Our goal in this study was to understand how dietary zinc supplementation affects thymopoiesis in aged mice. We hypothesized that impaired zinc status associated with aging would mediate the decline in thymic function and output and that restoring plasma zinc concentrations via zinc supplementation would improve thymopoiesis and thymic functions. In this study, groups of young (8 wk) and aged (22 mo) mice were fed a zinc-adequate (30 mg/kg zinc) or zinc-supplemented diet (300 mg/kg) for 25 d. Aged mice had impaired zinc status, with zinc supplementation restoring plasma zinc to a concentration not different from those of young male C57Bl/6 mice. Zinc supplementation in aged mice improved thymopoiesis, as assessed by increased total thymocyte numbers. In addition, improved thymic output was mediated in part by reducing the age-related accumulation of immature CD4(-)CD8(-)CD44(+)CD25(-) thymocytes, as well as by decreasing the expression of stem cell factor, a thymosuppressive cytokine. Taken together, our results showed that in mice, zinc supplementation can reverse some age-related thymic defects and may be of considerable benefit in improving immune function and overall health in elderly populations.
Content may be subject to copyright.
The Journal of Nutrition
Nutritional Immunology
Zinc Supplementation Increases Zinc Status and
Thymopoiesis in Aged Mice
1,2
Carmen P. Wong,
3
Yang Song,
3
Valerie D. Elias,
3
Kathy R. Magnusson,
4
and Emily Ho
3,5
*
3
Department of Nutrition and Exercise Sciences, Oregon State University, OR 97331; and
4
Department of Biomedical Sciences, and
5
Linus Pauling Institute, Oregon State University, Corvallis, OR 97331
Abstract
The age-related decline in lymphocyte development and function coincides with impaired zinc status in the elderly. Thymic
involution and reduced immune responsiveness are classic hallmarks of both aging and zinc deficiency, resulting in
decreased host defense and an increased susceptibility to infections. Thus, compromised zinc status associated with
aging may be an important contributing factor in reduced thymopoiesis and impaired immune functions. Our goal in this
study was to understand how dietary zinc supplementation affects thymopoiesis in aged mice. We hypothesized that
impaired zinc status associated with aging would mediate the decline in thymic function and output and that restoring
plasma zinc concentrations via zinc supplementation would improve thymopoiesis and thymic functions. In this study,
groups of young (8 wk) and aged (22 mo) mice were fed a zinc-adequate (30 mg/kg zinc) or zinc-supplemented diet (300
mg/kg) for 25 d. Aged mice had impaired zinc status, with zinc supplementation restoring plasma zinc to a concentration
not different from those of young male C57Bl/6 mice. Zinc supplementation in aged mice improved thymopoiesis, as
assessed by increased total thymocyte numbers. In addition, improved thymic output was mediated in part by reducing
the age-related accumulation of immature CD4
2
CD8
2
CD44
+
CD25
2
thymocytes, as well as by decreasing the expression
of stem cell factor, a thymosuppressive cytokine. Taken together, our results showed that in mice, zinc supplementation
can reverse some age-related thymic defects and may be of considerable benefit in improving immune function and overall
health in elderly populations. J. Nutr. 139: 1393–1397, 2009.
Introduction
Age-related decline in immune function encompasses multiple
defects (1). Among them, thymic involution and reduced T-cell
production are one of the most recognized hallmarks of aging.
Whereas the age-dependent changes in thymus function have
been well described, factors important in controlling the process
remain to be fully elucidated. The progressive age-related decline
in thymic function and output coincides with reduced zinc status
and suppressed immune responses in the elderly population.
Thus, the reduced zinc status that occurs with aging may play an
important role in mediating reduced thymopoiesis and contribute
to a progressive decline in immune responsiveness. This culmi-
nates in a higher incidence of infection, cancer, and autoimmune
diseases with increasing age (2–4).
Zinc is a key component for the functions of numerous
proteins and is an essential micronutrient required for numerous
cellular processes. In particular, zinc is necessary for the normal
development and function of the immune system (5). Alterations
in dietary zinc intake, zinc uptake, retention, or secretion can
lead to zinc deficiency and affect zinc-dependent functions. Zinc
deficiency can significantly depress immune response and impair
host defense (6). Zinc homeostasis is critically involved in the
signaling events in immune cells and changes in zinc status affect
multiple immune cell types involved in both innate and adap-
tive immunity (7–9). Severe zinc deficiency dramatically affects
the development of the immune system, resulting in immune
dysfunction. Hallmarks of severe zinc deficiency include thymic
involution, lymphopenia, and accelerated apoptosis in lympho-
cytes. One population particularly at risk for zinc deficiency is
the elderly, who have impaired zinc absorption and reduced
dietary intake (10,11). Elderly patients with reduced zinc status
have higher frequencies of infections (12) and reduced immune
responses to vaccinations (13–15). On the other hand, restoring
normal zinc levels in individuals with low zinc status via zinc
supplementation can improve T-cell–mediated functions and
decrease the incidence of infections in the elderly (16–18).
Zinc supplementation has been shown to reverse thymus
involution in aged mice (19,20). In human studies, zinc supple-
mentation improved immune functions in the elderly. However,
the mechanism of how this is accomplished is currently unclear.
In particular, whether improving zinc status through zinc supple-
mentation would improve thymopoiesis and thymic functions in
aged individuals remains to be investigated. We studied the
1
Supported by Oregon Agricultural Experiment Station (OR00735) and the
Environmental Health Science Center at Oregon State University (NIEHS P30
ES00210) and Linus Pauling Institute Pilot Grant Program.
2
Author disclosures: C. P. Wong, Y. Song, V. D. Elias, K. R. Magnusson, and
E. Ho, no conflicts of interest.
* To whom correspondence should be addressed. E-mail: emily.ho@
oregonstate.edu.
0022-3166/08 $8.00 ã2009 American Society for Nutrition.
Manuscript received February 19, 2009. Initial review completed February 26, 2009. Revision accepted May 5, 2009. 1393
First published online May 27, 2009; doi:10.3945/jn.109.106021.
effects of zinc supplementation on thymic development and
function in aged mice. We hypothesized that the impaired zinc
status of the elderly plays an important role in the age-related
decline in thymopoiesis and that zinc supplementation of an
aged population would improve zinc status and have direct
effects on thymic health by reversing age-related defects in the
thymus.
Materials and Methods
Mice, diets, and study design. Young (8 wk) C57Bl/6 male mice were
purchased from Jackson Laboratory. Aged (22 mo) C57Bl/6 male mice
were purchased from the National Institute on Aging. Mice were housed
in stainless steel suspended cages in a temperature- and humidity-
controlled environment and randomly assigned to either a zinc-adequate
(ZA)
6
diet containing 30 mg/kg zinc or a zinc-supplemented (ZS) diet
containing 10 times the zinc concentration (300 mg/kg zinc) of the ZA
diet that was previously shown to be well tolerated (21). Purified diets
were purchased from Research Diets and were custom prepared using an
egg white-based AIN-93G diet with zinc provided as zinc carbonate
(Table 1) (22). Mice were fed the assigned diets for 25 d and consumed
food and water ad libitum. The dietary intakes and body weights of all
mice were monitored throughout the study. At the termination of the
experiments, mice were killed by CO
2
asphyxiation. Blood was collected
for plasma isolation. Thymus from each mouse was collected and made
into single cell suspensions. Cell suspensions were passed through a 70-
mm cell strainer to remove cell debris and prepared for cell counting and
flow cytometry analysis. The animal protocol was approved by the
Oregon State University Institutional Laboratory Animal Care and Use
Committee.
Plasma zinc concentrations. Plasma zinc concentrations were mea-
sured using inductively coupled plasma-optical emission spectroscopy
as previously described, with minor modification (23). Briefly, plasma
samples (100 mL) were added to 1 mL 70% ultrapure nitric acid and
incubated overnight. Incubated samples were diluted with chelex-treated
nanopure water to a final concentration of 7% nitric acid, centrifuged
at 3000 3g; 1 min at 258C, and analyzed using the Prodigy High
Dispersion inductively coupled plasma-optical emission spectroscopy
instrument (Teledyne Leeman Labs) against known standards.
Cell counts and flow cytometry analysis. Thymocyte numbers were
determined using the Z1 Coulter Particle counter (Beckman Coulter).
After counting, one-half of the thymocytes were used for RNA isolation
and the remaining thymocytes were used in flow cytometry. For flow
cytometry analysis, thymocytes were resuspended in flow cytometry
buffer (PBS, 2% fetal bovine serum, 1 mmol/L EDTA). Cells were
incubated with CD4-FITC, CD25-PE, CD44-PerCP-Cy5.5, and CD8-
APC for 30 min on ice in the dark. All antibodies were purchased from
eBioscience. After extensive washing, cells were resuspended in buffer
for flow cytometry acquisition and analysis. A minimum of 300,000
events in the lymphocyte gate were collected. Data were acquired using
FACSCalibur (BD Biosciences). Data analyses were performed using
Summit software (DakoCytomation).
RNA isolation, cDNA synthesis, and real-time quantitative PCR.
Total RNA from thymocytes was isolated using Trizol reagent
(Invitrogen). One microgram of total RNA was reverse transcribed
into cDNA using SuperScript III First-Strand Synthesis SuperMix for
quantitative real-time PCR (Invitrogen). Real-time PCR was performed
using primers specific for mouse stem cell factor (SCF) (forward:
59-CAACTGCTCCTATTTAATCCTC-39, reverse: 59-TGTATTACCA-
TATCTCGTAGCC-39), interleukin-7 (IL-7) (forward: 59-CTAACAG-
TATCACAAGGCACAC-39, reverse: 59-TCAACCTCTCCAAGTATAT-
GAACC-39), or 18S ribosomal RNA (forward: 59-CCGCAGCTAG-
GAATAATGGAAT-39, reverse: 59-CGAACCTCCGACTTTCGTTCT-
39). Real-time PCR were performed using DyNAmo HS SYBR Green
qPCR kit (New England Biolabs). Gene copies were determined using
the standard curve method. A standard curve was generated from serial
dilutions of purified plasmid DNA that encoded for each gene of interest.
Data represent the copy number of the gene of interest normalized to the
copy number of the 18S ribosomal RNA housekeeping gene in individual
mice.
Statistical analysis. Data are reported as means 6SEM. The main
effects of age and dietary zinc and their interaction were analyzed using
2-way ANOVA, followed by the Bonferroni post hoc test when the
interaction was significant. Where necessary, data were log transformed
to correct for unequal variances prior to statistical analyses. Non-
transformed data are shown in tables and figures. All analyses were
performed using GraphPad Prism version 4.01. Significance was defined
as P,0.05.
Results
Plasma zinc concentration. Both dietary zinc and age affected
zinc status as assessed by the plasma zinc concentration (Fig.
1A). It was lower in aged mice than in young mice and was
greater in ZS mice than ZA mice. The greater plasma zinc in the
ZS mice was not due to differences in food intake or body weight
gain; these variables did not differ between ZS and ZA mice of
either age (data not shown).
Total and double negative thymocyte numbers. Dietary
zinc, age, and their interaction affected the number of thymo-
cytes (Fig. 1B). As expected, aged mice had fewer thymocytes
than young mice. Zinc supplementation did not alter thymocyte
numbers in young mice but resulted in a significant 52% increase
in thymocytes in aged mice.
We evaluated whether zinc status affected thymocyte devel-
opment in young and aged mice. Thymocyte maturation is
divided into 4 main differentiation stages based primarily on the
TABLE 1 Diet composition
1
ZA diet ZS diet
g/kg
Egg white, spray dried 203 203
L-Histidine 0 0.04
Corn starch 494.5 494.5
Maltodextrin 10 35 35
Sucrose 100 100
Cellulose 50 50
Soybean oil 70 70
t-Butylhydroquinone 0.014 0.014
Mineral mix S19409 (no added Ca, P, K, or Zn)
2
77
Potassium phosphate, monobasic 6.86 6.86
Calcium carbonate 8 8
Potassium citrate 2.48 2.48
Calcium phosphate 6 6
Vitamin mix V10037
2
10 10
Biotin, 1% 0.4 0.4
Choline bitartrate 2.5 2.5
Zinc carbonate (52.1% zinc) 0.058 0.58
1
Final zinc concentration is 30 mg/kg zinc in the ZA diet and 300 mg/kg zinc in the ZS
diet.
2
Mineral and vitamin mix as previously described (22).
6
Abbreviations used: DN, double negative; DP, double positive; IL-7,
interleukin-7; SP, single positive; SCF, stem cell factor; ZA, zinc adequate; ZS,
zinc supplemented.
1394 Wong et al.
expression of CD4 and CD8. The earliest immature thymocytes
are double negative (DN) thymocytes (CD4
2
CD8
2
), which tran-
sition to immature double positive (DP) (CD4
+
CD8
+
) thymo-
cytes, and finally differentiate into mature single positive (SP)
CD4
+
CD8
2
and CD8
+
CD4
2
thymocytes that exit the thymus
and enter the bloodstream (Fig. 2A). Dietary zinc and age, but
not their interaction, specifically affected DN thymocytes; there
were more in aged mice than in young mice (Table 2). Zinc
supplementation resulted in a lower frequency of DN thymo-
cytes compared with ZA mice. DP and SP thymocytes were not
affected by dietary zinc in aged or young mice.
DN1 thymocyte subsets. The development of DN thymocytes
can be further divided into 4 distinct maturation steps based on
the expression of CD44 and CD25, namely DN1 (CD44
+
CD25
2
),
DN2 (CD44
+
CD25
+
), DN3 (CD44
2
CD25
+
), and DN4
(CD44
2
CD25
2
) (24). During aging, there is a specific blockade
in thymocyte development that prevents DN1-DN2 transition,
resulting in the accumulation of DN1 thymocytes (Fig. 2A)
(25,26). Dietary zinc and age, but not their interaction, specif-
ically affected DN1 thymocytes (Fig. 2B). As expected, aged
mice had a higher frequency of the DN1 subset than young mice.
Zinc supplementation reduced the proportion of DN1 thymo-
cytes compared with ZA mice.
Thymic cytokines. Dysregulation of various thymic cytokines,
including SCF and IL-7, have been associated with thymic
atrophy and the suppression of thymopoiesis during aging (27).
Dietary zinc, age, and their interaction affected thymic SCF
expression (Fig. 1C). Specifically, SCF expression was greater in
the thymus of aged mice than in young mice. Zinc supplemen-
tation in young mice did not alter SFC expression but signifi-
cantly reduced SCF expression in aged mice. Neither age nor
zinc supplementation affected thymic IL-7 expression (data not
shown).
Discussion
There is a grow ing body of evidence tha t suggests that the impair ed
zinc status associated with aging may directly be involved in age-
related immunological decline. Thus, it has been postulated
that zinc supplementation to restore zinc levels in aged individ-
uals may have beneficial effects on immune health. This study
FIGURE 1 Plasma zinc concentration (A), thymocyte number (B),
and thymic SCF expression (C) in young and aged male C57Bl/6 mice
fed ZA or ZS diets for 25 d. Values are means 6SEM, n=8.
*Different from corresponding ZA, P,0.001. NS, Nonsignificant, P.
0.05.
FIGURE 2 Flow cytometry analysis
of DN (CD4
2
CD8
2
), DP (CD4
+
CD8
+
),
and SP (CD4
+
CD8
2
and CD8
+
CD4
2
)
thymocyte cell populations from
young and aged male C57Bl/6 mice
fed ZA or ZS diets for 25 d. DN cell
population was further analyzed to
determine the frequency of DN1
(CD44
+
CD25
2
), DN2 (CD44
+
CD25
+
),
DN3 (CD44
2
CD25
+
), and DN4
(CD44
2
CD25
2
). Representative flow
cytometry data (A) and mean DN1
frequency (B) are shown. Values are
means 6SEM, n= 8. NS, Nonsignif-
icant, P.0.05.
TABLE 2 CD4/CD8 thymocyte distribution in young and aged
male C57Bl/6 mice fed ZA or ZS diets for 25 d
1
Young Aged ANOVA
ZA ZS ZA ZS Diet Age Interaction
%P-values
DN 2.8 60.7 2.1 60.2 8.2 62.5 2.8 60.2 0.042 0.004 0.183
DP 88.9 60.5 88.0 60.5 81.2 63.6 88.2 60.7 0.162 0.098 0.081
CD4 SP 6.6 60.7 8.0 60.3 8.2 60.8 7.0 60.4 0.908 0.605 0.075
CD8 SP 1.6 60.1 2.0 60.2 2.3 60.3 1.9 60.1 0.968 0.163 0.061
1
Values are means 6SEM, n=8.
Zinc status and thymopoiesis in aged mice 1395
demonstrated that zinc status is impaired in aged mice and could
be restored to levels comparable to young mice by dietary zinc
supplementation. Moreover, our results indicated that zinc
supplementation partially reversed the thymic defects associ-
ated with aging. In particular, zinc supplementation increased
total thymocyte numbers in aged mice, in part by reversing the
age-related accumulation of DN1 thymocytes and reducing the
age-related elevated expression of the thymosuppressive cyto-
kine, SCF. These data were consistent with our hypothesis that
zinc status is an important variable in determining overall
thymic health during the aging process. To our knowledge, this is
the first report to directly examine the effects of zinc supplemen-
tation on thymocyte development and function during aging.
Overall, our data suggest that impaired zinc status associated
with the aging process may play an important role in the age-
related decline in thymopoiesis.
Immunosenescence, the age-related progressive decline in
lymphocyte development and function, coincides with impaired
zinc status in the elderly. Involution of the thymus and reduced
immune responsiveness are classic hallmarks shared by aging
and zinc deficiency. In particular, severe zinc deficiency results in
a decline of CD4
+
CD8
+
pre-T-cells in the thymus (5). Disruption
of the hypothalamus-pituitary-adrenal-axis leading to enhanced
circulating corticosterone concentrations has been postulated to
play an important role in zinc-deficiency related dysfunction
(28). Elevated corticosterone levels are also apparent in aging
and may also contribute to immunosenescence (29). Whether
alteration in corticosterone levels with age is directly related to
alterations in zinc status is currently unknown. Decreases in zinc
uptake and absorption increase the risk of zinc deficiency in the
elderly (10,11). Murine models have been used to study the
various effects of aging, including age-associated decline in zinc
status (19,30). In agreement with others, our data demonstrated
that aged C57Bl/6 mice, despite being fed a ZA diet, had a lower
plasma zinc concentration than young mice (Fig. 1A). This
suggested that aged mice, similar to humans, had impaired zinc
uptake/absorption. In addition, the elderly are more prone to
consuming inadequate levels of zinc in their diets. From the
NHANES III data, it is estimated that 12% of the U.S.
population does not consume the current Average Estimated
Requirement for zinc, but this number escalates to 35–45% in
people over the age of 60 y (31). Because a high proportion of
the elderly population (.60 y) consumes a low-zinc diet, there is
likely a synergy between low dietary zinc intake and the age-
related decline in zinc status that leads to immune dysfunction.
Our studies demonstrate that zinc supplementation of aged mice
restored plasma zinc to levels comparable to ZA young mice and
partially restored age-related alterations in thymopoiesis; thus, it
is likely that compromised zinc status only partially accounts for
the age-related decline in immune function. Understanding these
interactions is an important area of future research with
important public health implications and highlights the potential
need for higher zinc requirements in an elderly population.
The differentiation, selection, and output of mature naı
¨ve T-
cells take place in the thymus. The earliest immature thymocytes
are bone marrow-derived precursors that are negative for both
CD4 and CD8 (DN thymocytes). DN thymocytes are further
divided in 4 distinct subsets (DN1–4) based on the expression
of CD44 and CD25 (24). The frequency of immature DN1
thymocytes has been shown to be significantly increased in
aged mice, suggesting an age-dependent defect or blockade in
DN1-DN2 transition during thymocyte maturation (25,26). As
a consequence, a reduced number of thymocytes are available
for maturation and the output of naı
¨ve T-cells that exit the
thymus is reduced. Although zinc supplementation did not fully
restore thymocyte numbers to those of young mice, it neverthe-
less indicated that improving zinc status in aged mice could
enhance thymopoiesis by partially reversing involution of the
thymus (Fig. 1B). Our data further suggested that the increase in
thymocyte numbers was mediated in part by reducing the
accumulation of DN1 thymocytes and relieving the age-specific
DN1-DN2 block during thymopoiesis (Table 1; Fig. 2).
To date, the precise mechanism of age-induced thymic atrophy
remains unclear. However, it likely involves changes in the
thymic microenvironment, including the dysregulation of thy-
mic cytokines, the loss of thymic epithelium, and/or changes in
T-cell progenitors (1). Dysregulation of thymotrophic as well as
thymosuppressive cytokines have been proposed to contribute to
age-associated thymic involution (27). In particular, elevated
expression of thymosuppressive cytokines such as SCF has been
observed in aged thymus and administration of SCF into young
mice induced acute thymic atrophy in vivo. Although the precise
mechanism by which SCF affects thymopoiesis remains to be
elucidated, we showed that zinc supplementation in aged mice
was associated with a significantly reduced expression of SCF
in the thymus (Fig. 1C). Interestingly, dietary supplementation
of zinc similarly decreased expression of SCF in the small
intestine in vivo (32). In addition to the restoration of normal
expression of SCF in aged mice, zinc supplementation likely
affects thymopoiesis via additional mechanisms. For example,
dietary zinc may modulate the expression of genes involved in
zinc homeostasis. Our preliminary survey of zinc transporters
in the thymus revealed age-specific differences in zinc trans-
porter expression (data not shown). In addition, Moore et al.
(33) reported that the expressions of a number of genes were
modulated in the thymus of zinc-deficient and zinc-supplemented
young mice. Interestingly, genes differentially expressed with
age, including various thymic cytokines (27) and zinc trans-
porters (C. P. Wong and E. Ho, unpublished data), were not
among the list of differentially expressed genes identified in this
murine model of severe zinc deficiency. However, in this model,
p56
lck
, a lymphocyte-specific tyrosine kinase important in the
selection and maturation of thymocytes, was upregulated with
zinc deficiency. This could be an important target for examina-
tion in future studies. Other studies have also found increased
p56
lck
with zinc deficiency but without effects on T-cell matura-
tion (34). It is possible that alterations found with age-related
decline in zinc status are not fully comparable to young growing
mice fed a severely zinc-deficient diet (,1 mg/kg zinc). Both the
severity of zinc deficiency and the physiological response to
zinc status may differ. At the same time, the effects of zinc
supplementation likely have differential effects in young and
aged mice. This is evidenced in our findings that the effect of zinc
supplementation on SCF expression was exclusively observed in
aged mice. Further study is needed to characterize the response
of the immune system to nutritional status in the growing young,
adult, and aged animals. The interactions among nutrition, age,
maturity/decline of the immune system are important to address
to be able to make appropriate nutrient intake recommendations
across the lifespan. Future studies will focus on providing an in-
depth characterization of the differential expression of genes in
the thymus with age and examine how zinc status influences the
expression of other thymic cytokines as well as genes involved
in zinc homeostasis, such as metallothionein and zinc trans-
porters. We will further explore how zinc status and age influence
thymopoiesis. Specifically, it will be of great interest to study
how diet and age interact and affect cell proliferation and/or
apoptosis during thymocyte development.
1396 Wong et al.
In summary, we showed that zinc supplementation improved
impaired zinc status and thymopoiesis in aged mice. Reversing
age-related thymic involution and enhancing thymic develop-
ment and function would be of considerable benefit to improve
overall immune function in aged individuals. In particular, the
elderly are at increased risk of complications and death from
infections such as influenza. Improving thymic development and
function in the elderly should lead to an enhancement in immune
responsiveness and translate to a reduction in complications and
mortality from infections. Our study showed that zinc supple-
mentation can serve as a therapeutic agent in improving thymic
health and provided the foundation for future studies to further
investigate the mechanisms as well as therapeutic effects of zinc
supplementation in reversing thymic involution and in improv-
ing immune response in the elderly.
Literature Cited
1. Gruver AL, Hudson LL, Sempowski GD. Immunosenescence of ageing.
J Pathol. 2007;211:144–56.
2. Ershler WB, Longo DL. Aging and cancer: issues of basic and clinical
science. J Natl Cancer Inst. 1997;89:1489–97.
3. Louria DB, Sen P, Sherer CB, Farrer WE. Infections in older patients: a
systematic clinical approach. Geriatrics. 1993;48:28–34.
4. Boren E, Gershwin ME. Inflamm-aging: autoimmunity, and the
immune-risk phenotype. Autoimmun Rev. 2004;3:401–6.
5. Fraker PJ, King LE. Reprogramming of the immune system during zinc
deficiency. Annu Rev Nutr. 2004;24:277–98.
6. Fischer Walker C, Black RE. Zinc and the risk for infectious disease.
Annu Rev Nutr. 2004;24:255–75.
7. Ibs KH, Rink L. Zinc-altered immune function. J Nutr. 2003;133:
S1452–6.
8. Kitamura H, Morikawa H, Kamon H, Iguchi M, Hojyo S, Fukada T,
Yamashita S, Kaisho T, Akira S, et al. Toll-like receptor-mediated
regulation of zinc homeostasis influences dendritic cell function. Nat
Immunol. 2006;7:971–7.
9. Rink L, Haase H. Zinc homeostasis and immunity. Trends Immunol.
2006;28:1–4. Epub 2006 Nov 28.
10. Fairweather-Tait SJ, Harvey LJ, Ford D. Does ageing affect
zinc homeostasis and dietary requirements? Exp Gerontol. 2008;43:
382–8.
11. Haase H, Mocchegiani E, Rink L. Correlation between zinc status and
immune function in the elderly. Biogerontology. 2006;7:421–8.
12. Mocchegiani E, Giacconi R, Muzzioli M, Cipriano C. Zinc, infections
and immunosenescence. Mech Ageing Dev. 2000;121:21–35.
13. Hannoun C, Megas F, Piercy J. Immunogenicity and protective efficacy
of influenza vaccination. Virus Res. 2004;103:133–8.
14. Deng Y, Jing Y, Campbell AE, Gravenstein S. Age-related impaired type
1 T cell responses to influenza: reduced activation ex vivo, decreased
expansion in CTL culture in vitro, and blunted response to influenza
vaccination in vivo in the elderly. J Immunol. 2004;172:3437–46.
15. Looney RJ, Hasan MS, Coffin D, Campbell D, Falsey AR, Kolassa J,
Agosti JM, Abraham GN, Evans TG. Hepatitis B immunization of
healthy elderly adults: relationship between naive CD4+ T cells and
primary immune response and evaluation of GM-CSF as an adjuvant.
J Clin Immunol. 2001;21:30–6.
16. Bogden JD, Oleske JM, Lavenhar MA, Munves EM, Kemp FW,
Bruening KS, Holding KJ, Denny TN, Guarino MA, et al. Effects of one
year of supplementation with zinc and other micronutrients on cellular
immunity in the elderly. J Am Coll Nutr. 1990;9:214–25.
17. Kajanachumpol S, Srisurapanon S, Supanit I, Roongpisuthipong C,
Apibal S. Effect of zinc supplementation on zinc status, copper status
and cellular immunity in elderly patients with diabetes mellitus. J Med
Assoc Thai. 1995;78:344–9.
18. Prasad AS, Beck FW, Bao B, Fitzgerald JT, Snell DC, Steinberg JD,
Cardozo LJ. Zinc supplementation decreases incidence of infections in
the elderly: effect of zinc on generation of cytokines and oxidative
stress. Am J Clin Nutr. 2007;85:837–44.
19. Mocchegiani E, Santarelli L, Muzzioli M, Fabris N. Reversibility of the
thymic involution and of age-related peripheral immune dysfunctions
by zinc supplementation in old mice. Int J Immunopharmacol. 1995;17:
703–18.
20. Sbarbati A, Mocchegiani E, Marzola P, Tibaldi A, Mannucci R,
Nicolato E, Osculati F. Effect of dietary supplementation with zinc
sulphate on the aging process: a study using high field intensity MRI and
chemical shift imaging. Biomed Pharmacother. 1998;52:454–8.
21. Ho E, Quan N, Tsai YH, Lai W, Bray TM. Dietary zinc supplemen-
tation inhibits NFkappaB activation and protects against chemically
induced diabetes in CD1 mice. Exp Biol Med (Maywood). 2001;
226:103–11.
22. Reeves PG. Components of the AIN-93 diets as improvements in the
AIN-76A diet. J Nutr. 1997;127:S838–41.
23. Verbanac D, Milin C, Domitrovic R, Giacometti J, Pantovic R, Ciganj
Z. Determination of standard zinc values in the intact tissues of mice by
ICP spectrometry. Biol Trace Elem Res. 1997;57:91–6.
24. Godfrey DI, Kennedy J, Suda T, Zlotnik A. A developmental pathway
involving four phenotypically and functionally distinct subsets of CD3–
CD4-CD8- triple-negative adult mouse thymocytes defined by CD44
and CD25 expression. J Immunol. 1993;150:4244–52.
25. Phillips JA, Brondstetter TI, English CA, Lee HE, Virts EL, Thoman
ML. IL-7 gene therapy in aging restores early thymopoiesis without
reversing involution. J Immunol. 2004;173:4867–74.
26. Thoman ML. The pattern of T lymphocyte differentiation is altered
during thymic involution. Mech Ageing Dev. 1995;82:155–70.
27. Sempowski GD, Hale LP, Sundy JS, Massey JM, Koup RA, Douek DC,
Patel DD, Haynes BF. Leukemia inhibitory factor, oncostatin M, IL-6,
and stem cell factor mRNA expression in human thymus increases with
age and is associated with thymic atrophy. J Immunol. 2000;164:
2180–7.
28. Fraker PJ, Osati-Ashtiani F, Wagner MA, King LE. Possible roles for
glucocorticoids and apoptosis in the suppression of lymphopoiesis
during zinc deficiency: a review. J Am Coll Nutr. 1995;14:11–7.
29. Qiao S, Chen L, Okret S, Jondal M. Age-related synthesis of gluco-
corticoids in thymocytes. Exp Cell Res. 2008;314:3027–35.
30. Dardenne M, Boukaiba N, Gagnerault MC, Homo-Delarche F,
Chappuis P, Lemonnier D, Savino W. Restoration of the thymus in
aging mice by in vivo zinc supplementation. Clin Immunol Immuno-
pathol. 1993;66:127–35.
31. Ervin RB, Kennedy-Stephenson J. Mineral intakes of elderly adult
supplement and non-supplement users in the third national health and
nutrition examination survey. J Nutr. 2002;132:3422–7.
32. Ou D, Li D, Cao Y, Li X, Yin J, Qiao S, Wu G. Dietary supplementation
with zinc oxide decreases expression of the stem cell factor in the small
intestine of weanling pigs. J Nutr Biochem. 2007;18:820–6.
33. Moore JB, Blanchard RK, Cousins RJ. Dietary zinc modulates gene
expression in murine thymus: results from a comprehensive differential
display screening. Proc Natl Acad Sci USA. 2003;100:3883–8.
34. Hosea HJ, Rector ES, Taylor CG. Zinc-deficient rats have fewer recent
thymic emigrant (CD90+) T lymphocytes in spleen and blood. J Nutr.
2003;133:4239–42.
Zinc status and thymopoiesis in aged mice 1397
... Checklist. Adequate zinc levels aligned with recommended zinc levels in AIN-93G rodent diets and marginal zinc deficiency and zinc supplementation models have been established previously [9,21]. Diets were formulated using a modified egg white-based AIN-93G diet wherein zinc was provided as zinc carbonate as previously described. ...
... Purified ZA, ZD, and ZS diets were purchased from Research Diets (New Brunswick, NJ). The ZD diet and ZS diet had previously been shown to result in reduced zinc status and increased zinc status in mice, respectively [21,22]. Food and water were provided ad libitum, and food intakes and body weights of all mice were monitored throughout the study. ...
Article
Full-text available
Older adult populations are at risk for zinc deficiency, which may predispose them to immune dysfunction and age-related chronic inflammation that drives myriad diseases and disorders. Recent work also implicates the gut microbiome in the onset and severity of age-related inflammation, indicating that dietary zinc status and the gut microbiome may interact to impact age-related host immunity. We hypothesize that age-related alterations in the gut microbiome contribute to the demonstrated zinc deficits in host zinc levels and increased inflammation. We tested this hypothesis with a multifactor two-part study design in a C57BL/6 mouse model. The two studies included young (2 month old) and aged (24 month old) mice fed either (1) a zinc adequate or zinc supplemented diet, or (2) a zinc adequate or marginal zinc deficient diet, respectively. Overall microbiome composition did not significantly change with zinc status; beta diversity was driven almost exclusively by age effects. Microbiome differences due to age are evident at all taxonomic levels, with more than half of all taxonomic units significantly different. Furthermore, we found 150 out of 186 genera were significantly different between the two age groups, with Bacteriodes and Parabacteroides being the primary taxa of young and old mice, respectively. These data suggest that modulating individual micronutrient concentrations does not lead to comprehensive microbiome shifts, but rather affects specific components of the gut microbiome. However, a phylogenetic agglomeration technique (ClaaTU) revealed phylogenetic clades that respond to modulation of dietary zinc status and inflammation state in an age-dependent manner. Collectively, these results suggest that a complex interplay exists between host age, gut microbiome composition, and dietary zinc status.
... Zinc deprivation in the controlled diet group did not affect serum zinc concentrations. This is consistent with the findings of Wong et al. (2009) and might be explained by the fact that, in contrast to the acute, highdose treatment, fewer micronutrients were administered to the mice for a longer period, so the serum levels could be saturated. We have also found differences in basal immobility values between the experiments assessing the effect of acute Zn treatment and the effects of the zinc-controlled diet. ...
Article
Full-text available
Background: As a member of the purinergic receptor family, divalent cation-regulated ionotropic P2X7 (P2rx7) plays a role in the pathophysiology of psychiatric disorders. This study aimed to investigate whether the effects of acute zinc administration and long-term zinc deprivation on depression-like behaviors in mice are mediated by P2X7 receptors. Methods: The antidepressant-like effect of elevated zinc level was studied using a single acute intraperitoneal injection in C57BL6/J wild-type and P2rx7 gene-deficient (P2rx7 −/−) young adult and elderly animals in the tail suspension test (TST) and the forced swim test (FST). In the long-term experiments, depression-like behavior caused by zinc deficiency was investigated with the continuous administration of zinc-reduced and control diets for 8 weeks, followed by the same behavioral tests. The actual change in zinc levels owing to the treatments was examined by assaying serum zinc levels. Changes in monoamine and brain-derived neurotrophic factor (BDNF) levels were measured from the hippocampus and prefrontal cortex brain areas by enzyme-linked immunosorbent assay and high-performance liquid chromatography, respectively. Results: A single acute zinc treatment increased the serum zinc level evoked antidepressant-like effect in both genotypes and age groups, except TST in elderly P2rx7 −/− animals, where no significant effect was detected. Likewise, the pro-depressant effect of zinc deprivation was observed in young adult mice in the FST and TST, which was alleviated in the case of the TST in the absence of functional P2X7 receptors. Among elderly mice, no pro-depressant effect was observed in P2rx7 −/− mice in either tests. Treatment and genotype changes in monoamine and BDNF levels were also detected in the hippocampi. Conclusion: Changes in zinc intake were associated with age-related changes in behavior in the TST and FST. The antidepressant-like effect of zinc is partially mediated by the P2X7 receptor.
... Восполнение пищевого дефицита цинка нормализует показатели апоптоза тимоцитов [21]. Прием цинка с питьевой водой восстанавливает массу и клеточность тимуса у старых мышей [22,23], а также повышает у них пролиферативный ответ тимоцитов на митогены и цитокины [24]. ...
Article
Full-text available
Цель исследования. Изучить влияние перорального приема цинка на функциональную активность тимоцитов при росте перевиваемой опухоли гепатомы 22а у мышей. Материалы и методы. Мыши линии C3HA, начиная с первого дня после подкожной инокуляции клеток сингенной гепатомы 22а, получали сульфат цинка с питьевой водой в концентрации 22 мкг/мл в течение трех недель. На 21 сут опухолевого роста животных выводили их эксперимента, извлекали тимусы, оценивали пролиферативную активность и апоптоз тимоцитов, а также содержание цинка в тимусе. Пролиферацию (распределение тимоцитов по стадиям клеточного цикла) изучали с помощью проточной цитометрии путем окрашивания DAPI. Для исследования апоптоза клетки окрашивали DAPI и YO-PRO. Содержание цинка в тимусе определяли с помощью атомно-абсорбционной спектрометрии. Результаты. На 21 сутки роста опухоли апоптоз тимоцитов увеличивался в 2,5 раза, а доля тимоцитов, находящихся в S фазе (фазе синтеза ДНК), снижалась в 1,8 раза. Апоптоз, главным образом, обнаруживали среди популяции двойных позитивных CD4+CD8+ тимоцитов – было отмечено увеличение в 3,2 раза по сравнению с контролем. Прием цинка нормализовал показатели пролиферативной активности (пролиферативный индекс и долю клеток в S фазе), а также снижал относительное содержание тимоцитов в состоянии апоптоза. Кроме того, прием соли цинка повышал содержание цинка в вилочковой железе. Заключение. Пероральный прием сульфата цинка вызывает торможение инволюции тимуса у мышей с гепатомой 22а и значительно улучшает показатели функциональной активности тимоцитов. У таких животных пролиферативная активность тимоцитов сохраняется на нормальном уровне, а показатели апоптоза существенно ниже, чем у животных, не получавших соль цинка. Проведенное исследование дает возможность считать пероральный прием соли цинка перспективным средством для разработки новых стратегий по восстановлению тимуса у онкологических больных.
... Nevertheless, the amount of zinc in diets labeled by manufactures as standard varies between 25 (our previous study), 50 (Altromin GmbH), and 70 (Harlan) mg Zn/kg. In contrast, the zinc content of a zinc supplemented diet is several hundred mg Zn/kg (e.g., 248 mg Zn/kg [43], 246 mg Zn/kg [44] or 300 mg Zn/kg [45]). ...
Article
Full-text available
The G-protein coupled receptor 39 (GPR39) is gaining increasing attention as a target for future drugs, yet there are gaps in the understanding of its pharmacology. Zinc is an endogenous agonist or an allosteric modulator, while TC-G 1008 is a synthetic, small molecule agonist. Zinc is also a positive allosteric modulator for the activity of TC-G 1008 at GPR39. Activation of GPR39 by TC-G 1008 facilitated the development of epileptogenesis in the pentylenetetrazole (PTZ)-induced kindling model of epilepsy. Congruently, TC-G 1008 decreased the seizure threshold in the maximal electroshock seizure threshold (MEST) test. Here, we investigated the effects of TC-G 1008 under the condition of zinc deficiency. Mice were fed a zinc-adequate diet (ZnA, 50 mg Zn/kg) or a zinc-deficient diet (ZnD, 3 mg Zn/kg) for 4 weeks. Following 4 weeks of dietary zinc restriction, TC-G 1008 was administered as a single dose and the MEST test was performed. Additional groups of mice began the PTZ-kindling model during which TC-G 1008 was administered repeatedly and the diet was continued. TC-G 1008 administered acutely decreased the seizure threshold in the MEST test in mice fed the ZnD diet but not in mice fed the ZnA diet. TC-G 1008 administered chronically increased the maximal seizure severity and the percentage of fully kindled mice in those fed the ZnA diet, but not in mice fed the ZnD diet. Our data showed that the amount of zinc in a diet is a factor contributing to the effects of TC-G 1008 in vivo.
... That elderly are often deficient in zinc suggesting a direct correlation between chronic inflammation, immunosenescence, and increased susceptibility to infections (Cabrera 2015). Studies have demonstrated that dietary supplementation with zinc can reverse several facets of immunosenescence as evident by increased naïve T-cell subset (Wong et al. , 2021, improved immune cell functions (Barnett et al. 2016;Varin et al. 2008), increased thymopoiesis (Wong et al. 2009), attenuated inflamm-aging , and modulated Th1/Th2 immune homeostasis (Uciechowski et al. 2008) during aging. In addition to zinc, copper (Giacconi et al. 2017;Malavolta et al. 2015) and iron (Handono et al. 2021;Macciò and Madeddu 2012) deficiency is also correlated with aging and health, including immunosenescence. ...
Article
Full-text available
Immunological aging is strongly associated with the observable deleterious effects of human aging. Our understanding of the causes, effects, and therapeutics of aging immune cells has long been considered within the sole purview of immunosenescence. However, it is being progressively realized that immunosenescence may not be the only determinant of immunological aging. The cellular senescence-centric theory of aging proposes a more fundamental and specific role of immune cells in regulating senescent cell (SC) burden in aging tissues that has augmented the notion of senescence immunotherapy. Now, in addition, several emerging studies are suggesting that cellular senescence itself may be prevalent in aging immune cells, and that senescent immune cells exhibiting characteristic markers of cellular senescence, similar to non-leucocyte cells, could be among the key drivers of various facets of physiological aging. The present review integrates the current knowledge related to immunosenescence and cellular senescence in immune cells per se, and aims at providing a cohesive overview of these two phenomena and their significance in immunity and aging. We present evidence and rationalize that understanding the extent and impact of cellular senescence in immune cells vis-à-vis immunosenescence is necessary for truly comprehending the notion of an ‘aged immune cell’. In addition, we also discuss the emerging significance of dietary factors such as phytochemicals, probiotic bacteria, fatty acids, and micronutrients as possible modulators of immunosenescence and cellular senescence. Evidence and opportunities related to nutritional bioactive components and immunological aging have been deliberated to augment potential nutrition-oriented immunotherapy during aging.
... IL-7 administration in healthy mice enhanced the peripheral T cell subsets [107]. Apart from the strategies outlined above, various murine research studies have demonstrated the importance of IL-21, IL-22, IL-7, and zinc in restoring normal thymopoiesis [179][180][181]. All these treatments show a potential hope to control thymic involution and improve immune outcomes in individuals with compromised immune status as in adults or patients. ...
Article
The thymus is a primary lymphoid organ, essential for the development of T-cells that will protect from invading pathogens, immune disorders, and cancer. The thymus decreases in size and cellularity with age referred to as thymus involution or atrophy. This involution causes decreased T-cell development and decreased naive T-cell emigration to the periphery, increased proportion of memory T cells, and a restricted, altered T-cell receptor (TCR) repertoire. The changes in composition and function of the circulating T cell pool as a result of thymic involution led to increased susceptibility to infectious diseases including the recent COVID and a higher risk for autoimmune disorders and cancers. Thymic involution consisting of both structural and functional loss of the thymus has a deleterious effect on T cell development, T cell selection, and tolerance. The mechanisms which act on the structural (cortex and medulla) matrix of the thymus, the gradual accumulation of genetic mutations, and altered gene expressions may lead to immunosenescence as a result of thymus involution. Understanding the molecular mechanisms behind thymic involution is critical for identifying diagnostic biomarkers and targets for treatment help to develop strategies to mitigate thymic involution-associated complications. This review is focused on the consequences of thymic involution in infections, immune disorders, and diseases, identifying potential checkpoints and potential approaches to sustain or restore the function of the thymus particularly in elderly and immune-compromised individuals.
Preprint
Full-text available
Older adult populations are at risk for elevated zinc deficiency, which may predispose them to immune dysfunction and age-related chronic inflammation that drives myriad diseases and disorders. Recent work also implicates the gut microbiome in the onset and severity of age-related inflammation, indicating that dietary zinc status and the gut microbiome may interact to impact age-related host immunity. We hypothesize that age-related alterations in the gut microbiome contribute to the demonstrated zinc deficits in host zinc levels and increased inflammation. We tested this hypothesis with a multifactor two-part study design in a C57BL/6 mouse model. The two studies included young (2 month old) and aged (24 month old) mice fed either (1) a zinc adequate or zinc supplemented diet, or (2) a zinc adequate or marginal zinc deficient diet, respectively. Overall microbiome composition did not significantly change with zinc status; beta diversity was driven almost exclusively by age effects. Microbiome differences due to age are evident at all taxonomic levels, with more than half of all taxonomic units significantly different at each. Furthermore, we found 150 out of 186 genera were significantly different between the two age groups, with Bacteriodes and Parabacteroides being the primary components of young and old mice, respectively. These data suggest that modulating individual micronutrient concentrations does not lead to comprehensive microbiome shifts, but rather affects specific components of the gut microbiome. However, a phylogenetic agglomeration technique (ClaaTU) revealed phylogenetic clades of microbiota that respond to modulation of dietary zinc status in an age-dependent manner. Collectively, these results suggest that there exists a complex interplay between host age, gut microbiome composition, and dietary zinc status.
Article
Full-text available
Prolonged lymphopenia represents a major clinical problem after cytoreductive therapies such as chemotherapy and the conditioning required for hematopoietic stem cell transplant (HSCT), contributing toward the risk of infections and malignant relapse. Restoration of T cell immunity is dependent on tissue regeneration in the thymus, the primary site of T cell development; although the capacity of the thymus to repair itself diminishes over lifespan. However, although boosting thymic function and T cell reconstitution is of considerable clinical importance, there are currently no approved therapies for treating lymphopenia. Here we found that Zinc (Zn), is critically important for both normal T cell development as well as repair after acute damage. Accumulated Zn in thymocytes during development was released into the extracellular milieu after HSCT conditioning, where it triggered regeneration by stimulating endothelial cell-production of BMP4 via the cell surface receptor GPR39. Dietary supplementation of Zn was sufficient to promote thymic function in a mouse model of allogeneic HSCT, including enhancing the number of recent thymic emigrants in circulation; although direct targeting of GPR39 with a small molecule agonist enhanced thymic function without the need for prior Zn accumulation in thymocytes. Together, these findings not only define an important pathway underlying tissue regeneration, but also offer an innovative preclinical approach to treat lymphopenia in HSCT recipients.
Article
Full-text available
With advanced ageing the zinc pool undergoes progressive reduction as shown by the low zinc plasma levels and the negative crude zinc balance, both in humans and in rodents. It has been suggested that such zinc deficiency might be involved in many age-related immunological dysfunctions, including thymic failure. The relevance of zinc for good functioning of the entire immune system is, at present, well documented. In particular, zinc is required to confer biological activity to one of the best-known thymic peptides, thymulin, which is responsible for cell-mediated immunity. In deep zinc deficiencies, in humans and other animals, the low thymulin levels are due not to a primary failure of the thymus, but to a reduced peripheral saturation of thymic hormones by zinc ions. In aged mice both a reduced peripheral saturation of the hormone and a decreased production by the thymus were present. Oral zinc supplementation in old mice (22 months old) for 1 month induced a complete recovery of crude zinc balance from negative (−1.82) to positive values (+1.47), similar to those of young animals (+1.67). A full recovery of thymic functions with a regrowth of the organ and a partial restoration of the peripheral immune efficiency, as measured by mitogen responsiveness (PHA and ConA) and natural killer cell (NK) activity, were observed after zinc supplementation. These findings clearly pin-point the relevance of zinc for immune efficiency and suggest that the age-related thymic involution and peripheral immunological dysfunctions are not intrinsic and irreversible events but are largely dependent on the altered zinc pool.
Article
Full-text available
Glucocorticoids (GCs) are primarily synthesized in the adrenal glands but an ectopic production has also been reported in the brain, the gastrointestinal tract and in thymic epithelial cells (TEC). Here we show that thymocytes express genes encoding for all enzymes required for de novo GC synthesis and produce the hormone as demonstrated by both a GC specific reporter assay and a corticosterone specific ELISA assay. Interestingly, GC synthesis is detectable in cells from young mice (4 weeks) and thereafter increases during aging (14-22 weeks) together with an increased gene expression of the rate-limiting enzymes StAR and CYP11A1. Hormone production occurred at a thymocyte differentiation stage characterized by being double positive for the CD4 and CD8 surface markers but was found to be unrelated to CD69 expression, a marker for thymocytes undergoing positive selection. No GC synthesis was found in resting or anti-CD3 activated CD4 and CD8 positive T cells isolated from the spleen. Thymocyte-derived GC had an anti-proliferative effect on a GR-transfected cell line and induced apoptosis in thymocytes. The age- and differentiation stage-related GC synthesis in thymocytes may play a role in the involution process that the thymus gland undergoes.
Article
The AIN-93 rodent diets were formulated to substitute for the previous version (AIN-76A) and to improve the performance of animals that consume them. They are called AIN-93G, formulated for growth, and AIN-93M, for maintenance. Major changes included substituting cornstarch for sucrose and soybean oil for corn oil and increasing the amount in order to supply both essential fatty acids (linoleic and linolenic). L-Cystine was substituted for DL-methionine to supplement the casein component. The mineral mix was reformulated to lower the amounts of phosphorus, manganese and chromium, to increase the amount of selenium, and to add molybdenum, silicon, fluoride, nickel, boron, lithium and vanadium. The amounts of vitamins E, K-1 and B-12 were increased over those in the AIN-76A vitamin mix. The AIN-93G diet contains 200 g of casein and 70 g of soybean oil/kg diet. The maintenance diet (AIN-93M) contains 140 g of casein and 40 g of soybean oil/kg diet. The 1993 diets have a better balance of essential nutrients than the 1976 diet and are better choices for studies with laboratory rodents.
Article
Infections may cause mortality in old age due to damaged immune responses. As zinc is required as a catalyst, structural (zinc fingers) and regulatory ion, it is involved in many biological functions, including immune responses. Low zinc ion bioavailability and impaired cell-mediated immunity are common in ageing and may be restored by physiological supplementation with zinc for 1–2 months, impacting upon morbidity and survival. This article reviews the role of zinc in immune efficacy during ageing, and also describes the main biochemical pathways involved in the role of zinc in resistance to infections in ageing in order to better understand the possible causes of immunosenescence.
Article
The objective of this study was to determine the effects of a year of Zn supplementation on Zn concentrations in circulating cells and on cellular immune functions in the elderly. Subjects, aged 60-89, were given a placebo, 15 mg Zn, or 100 mg Zn daily for 12 months. All subjects also received a multivitamin/mineral supplement that contained no additional Zn. Blood samples were drawn and immune functions assessed prior to and at 3, 6, 12, and 16 months after beginning Zn supplementation. Subject diets were also assessed at each visit. Dietary folate, pyridoxine, alpha-tocopherol, copper, zinc, and magnesium were consistently below recommended intakes. Although plasma Zn increased significantly in the 100 mg Zn treatment group, concentrations of Zn in erythrocytes, mononuclear cells, polymorphonuclear leukocytes, and platelets were not significantly increased by zinc supplementation. Natural killer cell activity was transiently enhanced by the 100 mg/day dose of Zn. There was a progressive improvement in delayed dermal hypersensitivity (DDH) and in lymphocyte proliferative responses to two mitogens; this may have been due to one or more components of the multivitamin/mineral supplement administered to all study subjects. The enhancement of DDH was significantly greater in the placebo group than in either zinc treatment group. Thus, zinc had a beneficial effect on one measure of cellular immune function while simultaneously having an adverse effect on another measure of cellular immunity.
Article
This preliminary study was to investigate the effects of zinc supplementation on Zn status, Cu status, serum macrominerals and lymphocyte subsets in elderly diabetic patients. The results of Zn supplementation can correct plasma Zn levels to normal values. However, this treatment did not affect the cellular Zn, copper status and serum macrominerals. Enhancement of the percentage of CD4 cells was observed after Zn therapy but had no effect on the percentage of CD8 cells and CD4/CD8 ratios. Our finding implicated that zinc supplementation might be useful to enhance the immune status in these patients.
Article
Thymic atrophy and lymphopenia are immunological hallmarks of many forms of malnutrition including deficiencies in zinc. Extreme thymic atrophy (70-80%) along with a 50% loss of splenocytes in mice maintained on a zinc deficient diet (ZD) for 30 days suggested that the deficiency might be altering lymphopoiesis or the production of new lymphocytes by the bone marrow. As shown herein, mice who were marginally zinc deficient being 72-75% the body weight of adequately fed controls, exhibited a 50% decline in pre B-cells and a 25% decline in immature B-cells. The mature B-cells of the marrow appeared fairly resistant to effects of suboptimal zinc intake. Interesting, this pattern was similar to results obtained by treating bone marrow cells with levels of glucocorticoids analogous to those found in nutritionally deficient rodents. Furthermore, these same concentrations of steroids were shown to induce significant levels of apoptosis or cell death among pre and immature B-cells which accounted for their declining numbers subsequent to exposure to glucocorticoid. In order to better ascertain the potential role of glucocorticoids generated during zinc deficiency on lymphopoietic processes, adrenalectomies were performed in an attempt to remove glucocorticoids from the equation. Subsequently, adrenalectomized and sham operated mice were placed on a ZD or zinc adequate diet (ZA). Levels of steroids at the time of sacrifice were elevated six fold in non-adrenalectomized ZD mice compared to ZD adrenalectomized mice. Removal of the adrenal gland protected the thymus of ZD mice from atrophy and also provided substantial protection of lymphopoietic processes.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
We have subdivided mouse CD4-CD8-CD3- triple-negative (TN) thymocytes into four subsets based upon expression of CD44 and CD25, including CD44+CD25-, CD44+CD25+, CD44-CD25+ and CD44-CD25-. Characterization of these cells revealed several features distinct to each subset, in particular the expression of high levels of c-kit (the receptor for stem cell factor) by CD44+CD25-TN and CD44+CD25+TN but not by CD44-CD25+TN and CD44-CD25-TN. The CD44+CD25+TN subset also included the IL-7 and stem cell factor-responsive cells, whereas only minimal responsiveness was observed by the CD44- populations. These subsets also showed differential cytokine production potential (CD44+CD25- > CD44+CD25+ > CD44-CD25+ > CD44-CD25-) after stimulation with calcium ionophore, PMA and IL-1. The repopulation potential of these subsets in 2-deoxyguanosine-treated fetal thymic lobes supports the following maturation sequence: CD44+CD25- -->CD44+CD25+ -->CD44-CD25+ -->CD44-CD25-. Furthermore, the sequence of progression from CD44+CD25+ to CD44-CD25+ cells was confirmed by their TCR beta-chain gene configuration. The former population exhibits germ-line TCR beta-chain configuration, whereas the latter subset shows a rearranged pattern.
Article
The increased risk of infection among older patients can be divided into three clinical categories: infections increased in incidence, infections showing higher case fatality rates, and infections that are clinically worse, primarily because of late recognition. Among infections that are increased in incidence, the most important by far are tuberculosis and pneumococcal pneumonia. Infections that show higher fatality rates include influenza and--again--pneumococcal pneumonia. Intra-abdominal infections (eg, cholecystitis and appendicitis) are often clinically worse in older patients due to late recognition and delay in surgical intervention.
Article
A multiparametric study of the thymus was performed in normal aging mice (12-15 months old) submitted to a mild oral zinc supplementation during 3-6 months as compared to age-matched control mice. First, this study demonstrated that in rodents, zinc levels are significantly reduced with aging and can be restored to values close to those observed in young animals after 6 months of zinc supplementation. Second, our data showed that oral zinc administration stimulates thymus growth and partially restores the microenvironmental as well as lymphoid compartments of the organ. Regarding thymic endocrine function, a significant increase in thymulin levels and a concomitant decrease in plasma thymulin inhibitors were observed, suggesting that the age-related decline of thymic function might at least partially be due to extrinsic factors, such as zinc deficiency. The total number of thymic lymphocytes was consistently increased, without significant changes in CD4/CD8 defined thymocyte subsets. Finally, structural changes of the thymus epithelium were also detected, including the disappearance of epithelial cysts frequently observed in old animals, reappearance of a normal pattern of the thymic epithelial cell network, and a decrease in the extracellular matrix network. Taken together, these data suggest that aging-related physiological zinc deficiency induces some relevant changes in thymus structure and function which can be partially corrected by a mild oral zinc supplementation.