Acta Scientific Nutritional Health (ASNH)(ISSN: 2582-1423)

Review Article Volume 6 Issue 8

Impact of Diet and Nutrition on Memory T Cell Development, Maintenance and Function in the Context of Healthy Immune System

Joyeta Ghosh1, Khusboo Singh2, Sudrita Roy Choudhury2*, Samarpita Koner2 and Neelanjana Basu2

1Assistant Professor, Department of Dietetics and Nutrition, NSHM Knowledge Campus-Kolkata, West Bengal India
2Research Scholar, Department of Dietetics and Nutrition, NSHM Knowledge Campus-Kolkata, West Bengal India

*Corresponding Author: Sudrita Roy Choudhury, Research Scholar, Department of Dietetics and Nutrition, NSHM Knowledge Campus-Kolkata, West Bengal India.

Received: July 04, 2022; Published: July 29, 2022

Abstract

A well-functioning immune system is censorious for permanence in today’s earth. The immune system must be persistently alert, keep track of intimation of danger or invasion. Optimum nutrition is always one crucial factor for every cell to function optimally, including cells in the immune system as well. Memory T cells are one rudimentary component of immunological memory, furnishing rapid and powerful host protection against secondary challenges. The diet and nutritional status of the host are two major regulators of T cell functioning and immune system. Present article will review literature considering the crucial impact of diet and nutrition on memory T cell development, maintenance and function in the context of healthy immune function. Caloric restriction without having undernutrition influence memory T cell functioning, while undernutrition or protein energy malnutrition or diet induced obesity predisposes T cell dysfunctionality. On one hand, undernutrition causes immunodeficiency (increased susceptibility to infection), whereas overnutrition/obesity results in inflammation due to increase in pro-inflammatory regulators. Gut dysbiosis also has a significant role in T cell biology and host fitness. In order to maintain a healthy gut microbiome proper dietary intervention are very crucial. Although more detailed research is needed in the current field to unfold the exact role of balanced diet in T cell development, maintenance and function in the context of healthy immune function.

 

Keywords: Diet; T Cells; Immune System; Microbiota; Nutrition

References

  1. Calder PC., et al. “Undernutrition, infection and immune function”. Nutrition Research Reviews 13 (2000): 3-29.
  2. Lee GY., et al. “The Role of Vitamin E in Immunity”. Nutrients 10 (2018): 1614.
  3. Kaech SM., et al. “Transcriptional control of effector and memory CD8+ T cell differentiation”. Nature Reviews Immunology 12 (2012): 749-761.
  4. Mueller SN., et al. “Tissue-resident memory T cells: Local specialists in immune defense”. Nature Reviews Immunology 16 (2016): 79-89.
  5. Collins N., et al. “The Bone Marrow Protects and Optimizes Immunological Memory during Dietary Restriction”. Cell 178 (2019): 1088-1101.
  6. Romagnani S. “T-cell subsets (Th1 versus Th2)”. Annals of Allergy, Asthma and Immunology (2000): 9-18.
  7. Zhu J., et al. “Differentiation of effector CD4 T cell populations”. Annual Review of Immunology 28 (2010): 445-489.
  8. Schroeder HW., et al. “Structure and function of immunoglobulins”. Journal of Allergy and Clinical Immunology 125 (2010) :41-52.
  9. Berin MC. “Mucosal antibodies in the regulation of tolerance and allergy to foods”. Seminars in Immunopathology 34 (2012): 633-642.
  10. Vazquez MI., et al. “B cells responses and cytokine production are regulated by their immune microenvironment”. Cytokine 74 (2015): 318-326.
  11. Saraiva M., et al. “The regulation of IL-10 production by immune cells”. Nature Reviews Immunology 10 (2010): 170-181.
  12. Calder PC., et al. “Dietary factors and low-grade inflammation in relation to overweight and obesity”. British Journal of Nutrition 106 (2011): 5-78.
  13. Calder PC., et al. “Health relevance of the modification of low-grade inflammation in aging (inflammaging) and the role of nutrition”. Ageing Research Reviews 40 (2017): 95-119.
  14. Sallusto F., et al. “Two subsets of memory T lymphocytes with distinct homing potentials and effector functions”. Nature 401 (1999): 708-712.
  15. Jameson SC., et al. “Understanding Subset Diversity in T Cell Memory”. Immunity 48 (2018): 214-226.
  16. Schenkel JM., et al. “Sensing and alarm function of resident memory CD8+ T cells”. Nature Immunology 14 (2013) :509-513.
  17. Ariotti S., et al. “T cell memory. Skin-resident memory CD8+ T cells trigger a state of tissue-wide pathogen alert”. Science 346 (2014): 101-105.
  18. Gattinoni L., et al. “A human memory T cell subset with stem cell-like properties”. Nature Medicine 17 (2011): 1290-1297.
  19. Bromley SK., et al. “Recirculating memory T cells are a unique subset of CD4+ T cells with a distinct phenotype and migratory pattern”. The Journal of Immunology 190 (2013): 970-976.
  20. Gerlach C., et al. “The Chemokine Receptor CX3CR1 Defines Three Antigen-Experienced CD8 T Cell Subsets with Distinct Roles in Immune Surveillance and Homeostasis”. Immunity 45 (2016): 1270-1284.
  21. Fonseca R., et al. “Developmental plasticity allows outside-in immune responses by resident memory T cells”. Nature Immunology 21 (2020) :412-421.
  22. Boyman O., et al. “Cytokines and T-cell homeostasis”. Current Opinion in Immunology 19 (2007): 320-326.
  23. Becker TC., et al. “Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells”. Journal of Experimental Medicine 195 (2002): 1541-1548.
  24. Schluns KS., et al. “Cytokine control of memory T-cell development and survival”. Nature Reviews Immunology 3 (2003) :269-279.
  25. Surah CD., et al. “Homeostasis of memory T cells”. Immunological Reviews 211 (2006): 154-163.
  26. Cui G., et al. “IL-7-Induced Glycerol Transport and TAG Synthesis Promotes Memory CD8+ T Cell Longevity”. Cell 161 (2015): 750-761.
  27. Pan Y., et al. “Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism”. Nature 543 (2017): 252-256.
  28. O’Sullivan D., et al. “Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development”. Immunity 41 (2014): 75-88.
  29. Pearce EL., et al. “Enhancing CD8 T-cell memory by modulating fatty acid metabolism”. Nature 460 (2009): 103-107.
  30. Raud B., et al. “Fatty acid metabolism in CD8+ T cell memory: Challenging current concepts”. Immunological Reviews 283 (2018): 213-231.
  31. Raud B., et al. “Etomoxir Actions on Regulatory and Memory T Cells Are Independent of Cpt1a-Mediated Fatty Acid Oxidation”. Cell Metabolism 28 (2018) :504-515.
  32. O’Sullivan D. “The metabolic spectrum of memory T cells”. Immunology and Cell Biology 97 (2019): 636-646.
  33. Nikolich-Zugich J., et al. “Mice and flies and monkeys too: Caloric restriction rejuvenates the aging immune system of non-human primates”. Experimental Gerontology 40 (2005) :884-893.
  34. Contreras NA., et al. “Calorie restriction induces reversible lymphopenia and lymphoid organ atrophy due to cell redistribution”. Geroscience 40 (2018): 279-291.
  35. Lee C., et al. “Dietary restriction with and without caloric restriction for healthy aging”. F1000 Research (2016).
  36. Brandhorst S., et al. “Fasting and Caloric Restriction in Cancer Prevention and Treatment”. Recent Results in Cancer Research 207 (2016): 241-266.
  37. Fontana L., et al. “Growth factors, nutrient signaling, and cardiovascular aging”. Circulation Research 110 (2012): 1139-1150.
  38. Lee C., et al. “Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy”. Science Translational Medicine 4 (2012): 124ra27.
  39. Mattson MP., et al. “Impact of intermittent fasting on health and disease processes”. Ageing Research Reviews 39 (2017): 46-58.
  40. Kalaany NY., et al. “Tumors with PI3K activation are resistant to dietary restriction”. Nature 458 (2009): 725-731.
  41. Robertson LT., et al. “Benefits of short-term dietary restriction in mammals”. Experimental Gerontology 48 (2013): 1043-1048.
  42. Meydani SN., et al. “Long-term moderate calorie restriction inhibits inflammation without impairing cell-mediated immunity: A randomized controlled trial in non-obese humans”. Aging (Albany NY) 8 (2016): 1416-1431.
  43. Nagai M., et al. “Fasting-Refeeding Impacts Immune Cell Dynamics and Mucosal Immune Responses”. Cell 178 (2019): 1072-1087.
  44. Jordan S., et al. “Dietary Intake Regulates the Circulating Inflammatory Monocyte Pool”. Cell 178 (2019): 1102-1114.e17.
  45. Goldberg EL., et al. “Bone Marrow: An Immunometabolic Refuge during Energy Depletion”. Cell Metab 30 (2019): 621-623.
  46. Besedovsky L., et al. “Endogenous glucocorticoid receptor signaling drives rhythmic changes in human T-cell subset numbers and the expression of the chemokine receptor CXCR4”. FASEB Journal 28 (2014): 67-75.
  47. Besedovsky L., et al. “Cortisol increases CXCR4 expression but does not affect CD62L and CCR7 levels on specific T cell subsets in humans”. American Journal of Physiology-Endocrinology And Metabolism 306 (2014): E1322-E1329.
  48. Ghosh MC., et al. “Dexamethasone augments CXCR4-mediated signaling in resting human T cells via the activation of the Src kinase Lck”. Blood 113 (2009): 575-584.
  49. Shimba A., et al. “Glucocorticoids Drive Diurnal Oscillations in T Cell Distribution and Responses by Inducing Interleukin-7 Receptor and CXCR4”. Immunity 48 (2018): 286-298.
  50. Cawthorn WP., et al. “Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction”. Cell Metabolism 20 (2014): 368-375.
  51. Scheller EL., et al. “Marrow Adipose Tissue: Trimming the Fat”. Trends in Endocrinology and Metabolism (2016): 392-403.
  52. MJ Devlin. "Why does starvation make bones fat?" American Journal of Human Biology (2011): 577-585.
  53. Devlin MJ., et al. “Caloric restriction leads to high marrow adiposity and low bone mass in growing mice”. Journal of Bone and Mineral Research: The Official Journal of the American Society for Bone and Mineral Research (2010): 2078-2088.
  54. Han SJ., et al. “White Adipose Tissue Is a Reservoir for Memory T Cells and Promotes Protective Memory Responses to Infection”. Immunity (2017): 1154-1168.
  55. Misumi I., et al. “Obesity Expands a Distinct Population of T Cells in Adipose Tissue and Increases Vulnerability to Infection”. Cell Reports (2019): 214-224.
  56. Masopust D., et al. “Preferential localization of effector memory cells in nonlymphoid tissue”. Science (2001): 2413-2417.
  57. Contreras NA., et al. “Life-long control of cytomegalovirus (CMV) by T resident memory cells in the adipose tissue results in inflammation and hyperglycemia”. PLOS Pathogens (2019).
  58. Bourke Claire D., et al. "Immune Dysfunction as a Cause and Consequence of Malnutrition”. Trends in Immunology (2016): 386-398.
  59. Acosta-Rodríguez VA., et al. “Mice under Caloric Restriction Self-Impose a Temporal Restriction of Food Intake as Revealed by an Automated Feeder System”. Cell Metabolism (2017): 267-277.
  60. Prendergast Andrew J. “Malnutrition and vaccination in developing countries”. Philosophical transactions of the Royal Society of London”. Series B, Biological Sciences1671 (2015): 20140141.
  61. Schaible Ulrich E and Stefan H E Kaufmann. “Malnutrition and infection: complex mechanisms and global impacts”. PLoS Medicine5 (2007): e115.
  62. Brown Eric M., et al. “Diet and specific microbial exposure trigger features of environmental enteropathy in a novel murine model”. Nature Communications 7806 (2015).
  63. Iyer Smita S., et al. “Protein energy malnutrition impairs homeostatic proliferation of memory CD8 T cells”. Journal of Immunology (Baltimore, Md.: 1950)1 (2012): 77-84.
  64. Hoang Truc., et al. “Protein energy malnutrition during vaccination has limited influence on vaccine efficacy but abolishes immunity if administered during Mycobacterium tuberculosis infection”. Infection and Immunity5 (2015): 2118-2126.
  65. Taylor Andrew K., et al. “Protein energy malnutrition decreases immunity and increases susceptibility to influenza infection in mice”. The Journal of Infectious Diseases3 (2013): 501-510.
  66. Chatraw JH., et al. “Diminished primary CD8 T cell response to viral infection during protein energy malnutrition in mice is due to changes in microenvironment and low numbers of viral-specific CD8 T cell precursors”. The Journal of Nutrition 138 (2008): 806-812.
  67. Youngblood Ben., et al. “Effector CD8 T cells dedifferentiate into long-lived memory cells”. Nature 7685 (2017): 404-409.
  68. Akondy Rama S., et al. “Origin and differentiation of human memory CD8 T cells after vaccination”. Nature7685 (2017): 362-367.
  69. Ma Eric H., et al. “Serine Is an Essential Metabolite for Effector T Cell Expansion”. Cell Metabolism2 (2017): 345-357.
  70. Roy Dominic G., et al. “Methionine Metabolism Shapes T Helper Cell Responses through Regulation of Epigenetic Reprogramming”. Cell Metabolism2 (2020): 250-266.e9.
  71. Geiger Roger., et al. “L-Arginine Modulates T Cell Metabolism and Enhances Survival and Anti-tumor Activity”. Cell3 (2016): 829-842.e13.
  72. Murali-Krishna., et al. “Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection”. Immunity (1998): 8: 177-187.
  73. Ahmed R., et al. “Immunological memory and protective immunity: understanding their relation”. Science 272 (1996): 54-60.
  74. Savy M., et al. “Landscape analysis of interactions between nutrition and vaccine responses in children”. The Journal of Nutrition 139 (2009): 2154S-2218S.
  75. Chinen J., et al. “Secondary immunodeficiencies, including HIV infection”. Journal of Allergy and Clinical Immunology 2 (2010): S195-S203.
  76. Jewett JF., et al. “Preventive health care for adults with HIV infection”. JAMA 269 (1993): 1144-1153.
  77. Iyer Smita S., et al. "Protein energy malnutrition impairs homeostatic proliferation of memory CD8 T cells”. The Journal of Immunology1 (2012): 77-84.
  78. Salimonu LS., et al. “Lymphocyte subpopulations and antibody levels in immunized malnourished children”. British Journal of Nutrition 48 (1982): 7-14.
  79. , et al. “Development of impaired cell-mediated immunity in mild and moderate malnutrition”. The American Journal of Clinical Nutrition 34 (1981): 68-77.
  80. Carr EL., et al. “Glutamine uptake and metabolism are coordinated by ERK/MAPK during T lymphocyte activation”. The Journal of Immunology 185 (2010): 1037-1044.
  81. Mathews JD., et al. “Protein supplementation and enhanced antibody-producing capacity in New Guinean school-children”. Lancet (1972): 2: 675-677.
  82. Chandra RK. "Numerical and functional deficiency in T helper cells in protein energy malnutrition”. Clinical and Experimental Immunology1 (1983): 126.
  83. CHANDRA RK. “Immunodeficiency in undernutrition and overnutrition”. Nutrition Reviews 39 (1981c): 225.
  84. CHANDRA RK. “Cell-mediated immunity in nutritional imbalance”. Federation Proceedings 39 (1980b): 3088.
  85. Pontzer Herman., et al. “Hunter-gatherer energetics and human obesity”. PloS One7 (2012): e40503.
  86. Ma Eric H., et al. “Metabolic Profiling Using Stable Isotope Tracing Reveals Distinct Patterns of Glucose Utilization by Physiologically Activated CD8+ T Cells”. Immunity5 (2019): 856-870.e5.
  87. Andersen Catherine J., et al. “Impact of Obesity and Metabolic Syndrome on Immunity”. Advances in Nutrition (Bethesda, Md.)1 (2016): 66-75.
  88. Honce Rebekah and Stacey Schultz-Cherry. “Impact of Obesity on Influenza A Virus Pathogenesis, Immune Response, and Evolution”. Frontiers in Immunology 10 (2019): 1071.
  89. Louie Janice K., et al. “A novel risk factor for a novel virus: obesity and 2009 pandemic influenza A (H1N1)”. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America3 (2011): 301-312.
  90. Morgan Oliver W., et al. “Morbid obesity as a risk factor for hospitalization and death due to 2009 pandemic influenza A (H1N1) disease”. PloS One3 (2010): e9694.
  91. Fezeu L., et al. “Obesity is associated with higher risk of intensive care unit admission and death in influenza A (H1N1) patients: a systematic review and meta-analysis”. Obesity Reviews: An Official Journal of the International Association for the Study of Obesity8 (2011): 653-659.
  92. Weber DJ., et al. “Obesity as a predictor of poor antibody response to hepatitis B plasma vaccine”. JAMA 22 (1985): 3187-3189.
  93. Eliakim Alon., et al. “Reduced tetanus antibody titers in overweight children”. Autoimmunity 2 (2006): 137-141.
  94. Sheridan PA., et al. “Obesity is associated with impaired immune response to influenza vaccination in humans”. International Journal of Obesity (2005)8 (2012): 1072-1077.
  95. Neidich SD., et al. “Increased risk of influenza among vaccinated adults who are obese”. International Journal of Obesity (2005)9 (2017): 1324-1330.
  96. Park Hye-Lim., et al. “Obesity-induced chronic inflammation is associated with the reduced efficacy of influenza vaccine”. Human Vaccines and Immunotherapeutics5 (2014): 1181-1186.
  97. Park Hye-Lim., et al. “Obesity-induced chronic inflammation is associated with the reduced efficacy of influenza vaccine”. Human Vaccines and Immunotherapeutics5 (2014): 1181-1186.
  98. Karlsson Erik A., et al. “Diet-induced obesity in mice reduces the maintenance of influenza-specific CD8+ memory T cells”. The Journal of Nutrition9 (2010): 1691-1697.
  99. Rebeles Jennifer., et al. “Obesity-Induced Changes in T-Cell Metabolism Are Associated with Impaired Memory T-Cell Response to Influenza and Are Not Reversed with Weight Loss”. The Journal of Infectious Diseases10 (2019): 1652-1661.
  100. Han Seong-Ji., et al. “White Adipose Tissue Is a Reservoir for Memory T Cells and Promotes Protective Memory Responses to Infection”. Immunity6 (2017): 1154-1168.e6.
  101. Misumi Ichiro., et al. “Obesity Expands a Distinct Population of T Cells in Adipose Tissue and Increases Vulnerability to Infection”. Cell Reports2 (2019): 514-524.e5.
  102. Macdonald Thomas T and Giovanni Monteleone. “Immunity, inflammation, and allergy in the gut”. Science (New York, N.Y.)5717 (2005): 1920-1925.
  103. Colin Hill., et al. “The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics”. Nature Reviews Gastroenterology and Hepatology (2014): 506-514.
  104. Gibson Glenn R., et al. “Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics”. Nature reviews”. Gastroenterology and Hepatology8 (2017): 491-502.
  105. Hansen Nina Wærling and Anette Sams. “The Microbiotic Highway to Health-New Perspective on Food Structure, Gut Microbiota, and Host Inflammation”. Nutrients 11 (2018): 1590.
  106. Bischoff Stephan C., et al. “Intestinal permeability--a new target for disease prevention and therapy”. BMC Gastroenterology 14 (2014): 189.
  107. Sassi Francesca., et al. “Vitamin D: Nutrient, Hormone, and Immunomodulator”. Nutrients 11 (2018): 1656.
  108. Kiewiet Mensiena BG., et al. “Immunomodulatory Protein Hydrolysates and Their Application”. Nutrients7 (2018): 904.
  109. Santiago-López Lourdes., et al. “Effect of Milk Fermented with Lactobacillus fermentum on the Inflammatory Response in Mice”. Nutrients8 (20108): 1039.
  110. McKeen Starin., et al. “Infant Complementary Feeding of Prebiotics for theMicrobiome and Immunity”. Nutrients 2 (2019): 364.
  111. Brown Eric M., et al. “Gut Microbiota Regulation of T Cells During Inflammation and Autoimmunity”. Annual Review of Immunology 37 (2019): 599-624.
  112. Misumi Ichiro., et al. “Obesity Expands a Distinct Population of T Cells in Adipose Tissue and Increases Vulnerability to Infection”. Cell Reports2 (2019): 514-524.e5.
  113. Gerriets Valerie A and Nancie J MacIver. “Role of T cells in malnutrition and obesity”. Frontiers in Immunology 5 (2014): 379.
  114. Brown Kirsty., et al. “Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease”. Nutrients 8 (2012): 1095-119.
  115. Brown Kirsty., et al. “Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease”. Nutrients 8 (2012): 1095-119.
  116. Sonnenburg, Erica D et al. “Diet-induced extinctions in the gut microbiota compound over generations”. Nature 7585 (2016): 212-215.
  117. Gentile Christopher L and Tiffany L Weir. “The gut microbiota at the intersection of diet and human health”. Science (New York, N.Y.)6416 (2018): 776-780.
  118. Sonnenburg Erica D and Justin L Sonnenburg. “Starving our microbial self: the deleterious consequences of a diet deficient in microbiota-accessible carbohydrates”. Cell Metabolism 5 (2014): 779-786.
  119. Kaptan Zülal., et al. “Long term consequences on spatial learning-memory of low-calorie diet during adolescence in female rats; hippocampal and prefrontal cortex BDNF level, expression of NeuN and cell proliferation in dentate gyrus”. Brain Research 1618 (2015): 194-204.
  120. Liu Zhigang., et al. “High-fat diet induces hepatic insulin resistance and impairment of synaptic plasticity”. PloS One5 (2015): e0128274.
  121. Chaplin David D. “Overview of the immune response”. The Journal of Allergy and Clinical Immunology2 (2010): S3-23.

Citation

Citation: Sudrita Roy Choudhury., et al. “Impact of Diet and Nutrition on Memory T Cell Development, Maintenance and Function in the Context of ealthy Immune System". Acta Scientific Nutritional Health 6.8 (2022): 142-154.

Copyright

Copyright: © 2022 Sudrita Roy Choudhury., et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.




Metrics

Acceptance rate30%
Acceptance to publication20-30 days
Impact Factor1.316

Indexed In





News and Events


  • Certification for Review
    Acta Scientific certifies the Editors/reviewers for their review done towards the assigned articles of the respective journals.
  • Submission Timeline for Upcoming Issue
    The last date for submission of articles for regular Issues is July 10, 2024.
  • Publication Certificate
    Authors will be issued a "Publication Certificate" as a mark of appreciation for publishing their work.
  • Best Article of the Issue
    The Editors will elect one Best Article after each issue release. The authors of this article will be provided with a certificate of "Best Article of the Issue"
  • Welcoming Article Submission
    Acta Scientific delightfully welcomes active researchers for submission of articles towards the upcoming issue of respective journals.

Contact US