IMMUNE DYSFUNCTION AND COGNITIVE DEFICIT IN STRESS AND PHYSIOLOGICAL AGING. PART II: NEW APPROACHES TO COGNITIVE DISORDER PREVENTION AND TREATMENT

Cover Page


Cite item

Full Text

Abstract

Long-term stress as well as physiological aging result in similar immunological and hormonal disturbances including hypothalamic-pituitaryadrenal) axis depletion, aberrant immune response (regulatory T-cells, Tregs, and Th17-lymphocyte accumulation) and decreased dehydroepiandrosterone synthesis both in the brain and in the adrenal glands. Since the main mechanisms of inflammation control, «prompt» (stress hormones) and «delayed» (Tregs), are broken, serum cytokine levels increase and become sufficient for blood-brain-barrier disruption. As a result peripheral cytokines penetrate into the brain where they begin to perform new functions. Structural and functional alterations of blood-brain-barrier as well as stress- (or age-) induced neuroinflammation promote influx of bone marrow derived dendritic cells and lymphocyte effectors into the brain parenchyma. Thereafter, mass intrusion of pro-inflammatory mediators and immune cells having a lot of specific targets alters the brain work that we can observe both in humans and in animal experiments. The concept of stressful cognitive dysfunction, which is under consideration in this review, allows picking out several therapeutic targets: 1) reduction of excessive Treg accumulation; 2) supporting hypothalamic-pituitary-adrenal axis and inflammatory reaction attenuation; 3) recovery of dehydroepiandrosterone level; 4) improvement of blood-brain-barrier function.

About the authors

A. L. Pikhal'skii

Research Centre for Medical Genetics, Moscow, Russian Federation

Email: info@gabrich.com
The authors have indicated they have no financial relationships relevant to this article to disclose. Russian Federation

G. V. Shmarina

Research Centre for Medical Genetics, Moscow

Author for correspondence.
Email: osugariver@yahoo.com
MD, leading research scientist of the Department of Cystic fibrosis of Research Centre of Medical Genetics (RCMG) Russian Federation

V. A. Aleshkin

Gabrichevsky Institute of Epidemiology and Microbiology, Moscow

Email: info@gabrich.com

PhD, professor, Director of G.N. Gabrichevskii Moscow Research Institute for Epidemiology and Microbiology

 

 

Russian Federation

References

  1. Bower J.E., Ganz P.A., Aziz N. Altered cortisol response to psychologic stress in breast cancer survivors with persistent fatigue. Psychosom. Med. 2005; 67: 277–280.
  2. Gaab J., Baumann S., Budnoik A., Gmünder H., Hottinger N., Ehlert U. Reduced reactivity and enhanced negative feedback sensitivity of the hypothalamus-pituitary-adrenal axis in chronic whiplash-associated disorder. Pain. 2005; 119: 219–224.
  3. Pukhalsky A., Shmarina G., Alioshkin V. The Number of Regulatory T Cells: Purs uit of the Golden Mean; in: Regulatory T Cells 2. S.R. Hayashi (ed.). Nova Science Publishers Inc. 2010. P. 261–268. ISBN: 978-1-61761-554-2.
  4. Kipnis J., Schwartz M. Controlled autoimmunity in CNS maintenance and repair: naturally occurring CD4+CD25+ regulatory T-Cells at the crossroads of health and disease. Neuromol. Med. 2005; 7: 197–206.
  5. Wolf S.A., Steiner B., Akpinarli A., Kammertoens T., Nassenstein C., Braun A., Blankenstein .T, Kempermann G. CD4- positive T lymphocytes provide a neuroimmunological link in the control of adult hippocampal neurogenesis. J. Immunol. 2009; 182: 3979–3984.
  6. Maninger N., Wolokowitz O.M., Reus V.I., Epel E.S., Mellon SH. Neurological and neuropsychiatric effects of dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEAS). Front. Neuroendocrinoil. 2009; 30: 65–91.
  7. Hazeldine J., Arlt W., Lord J.M. Dehydroepiandrosterone as a regulator of immune cell function. J. Steroid Biochem. Mol. Biol. 2010; 120: 127–136.
  8. Гончаров Н.П., Кация Г.В. В кн.: Гормон здоровья и долголетия. М.: АДАМАНТЪ. 2012. 159 с.
  9. Suzuki T., Suzuki N., Engleman E.G., Mizushima Y, Sakane T. Low serum levels of dehydroepiandrosterone may cause deficient IL-2 production by lymphocytes in patients with systemic lupus erythematosus (SLE). Clin. Exp. Immunol. 1995; 99: 251–255.
  10. Setoguchi R., Hori,S., Takahashi T., Sakaguchi S. Homeostatic maintenance of natural Foxp3+ CD25+ CD4+ regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J. Exp. Med. 2005; 201: 723–735.
  11. Darrasse-Jeze G., Deroubaix S., Mouquet H., Victora G.D., Eisenreich T., Yao K.H., Masilamani R.F., Dustin M.L., Rudensky A., Liu K., Nussenzweig M.C. Feedback control of regulatory T cell homeostasis by dendritic cells in vivo. J. Exp. Med. 2009; 206: 1853–1862.
  12. Tesar B.M., Du W., Shirali A.C., Walker W.E., Walker W.E., Shen H., Goldstein D.R. Aging augments IL-17 T-cell alloimmune responses. Am. J. Transplant. 2008; 9: 54–63.
  13. Morita T., Mizutani Y., Sawada M., Shimada A. Immunohistochemical and ultrastructural findings related to the blood-- brain barrier in the blood vessels of the cerebral white matter in aged dogs. J. Comp. Pathol. 2005; 133: 14–22.
  14. Stichel C.C., Luebbert H. Inflammatory processes in the aging mouse brain: participation of dendritic cells and T-cells. Neurobiol. Aging. 2007; 28: 1507–1521.
  15. Kaunzner U.W., Miller M.M., Gottfried-Blackmore A., Gal-Toth J., Felger J.C., McEwen B.S., Bulloch K. Accumulation of resident and peripheral dendritic cells in the aging CNS. Neurobiol. Aging. 2012; 33: 681–693.
  16. Trzonkovski P., Szmit E., Mysliwska J., Mysliwski A. CD4+CD25+ T regulatory cells inhibit cytotoxic activity of CTL and NK cells on humans-impact of immunosenescence. Clin. Immunol. 2006; 119: 307–316.
  17. Ha T-Y. The Role of Regulatory T Cells in Cancer. Immune Network. 2009; 9: 209–235.
  18. Holmén N., Lundgren A., Lundin S. Functional CD4+CD25high regulatory T cells are enriched in the colonic mucosa of patients with active ulcerative colitis and increase with disease activity. Inflamm. Bowel Dis. 2006; 12: 447–456.
  19. Lee J.H., Yu H.H., Wang L.C., Yang Y.H., Lin Y.T., Chiang B.L. The levels of CD4+CD25+ regulatory T cells in paediatric patients with allergic rhinitis and bronchial asthma. Clin. Exp. Immunol. 2007; 148: 53–63.
  20. Lane N., Robins R.A., Corne J., Fairclough L. Regulation in chronic obstructive pulmonary disease: the role of regulatory T-cells and Th17 cells. Clin. Sci. (Lond.). 2010; 119: 75–86.
  21. Han G.M., O’Neil-Andersen N.J., Zurier R.B., Lawrence D.A. CD4+CD25 high T cell numbers are enriched in the peripheral blood of patients with rheumatoid arthritis. Cell Immunol. 2008; 253: 92–101.
  22. Schwartz M., Kipnis J. Therapeutic T cell-based vaccination for neurodegenerative disorders: the role of CD4+CD25+ regulatory T cells. Ann. N.Y. Acad. Sci. 2005; 1051: 701–708.
  23. Balcome S., Park S., Quirk Dorr D.R., Hafner L., Phillips L., Tretyakova N. Adenine-containing DNA-DNA crosslinks of antitumor nitrogen mustards. Chem. Res. Toxicol. 2004; 17: 950-962.
  24. Shirai H., Poetsch A.R., Gunji A., Maeda D., Fujimori H., Fujihara H., Yoshida T., Ogino H., Masutani M. PARG dysfunction enhances DNA double strand break formation in S-phase after alkylation DNA damage and augments different cell death pathways. Cell Death Dis. 2013; 4: e656.
  25. Kuppner M.C., Bleifuss E., Noessner E., Mocikat R., Hesler C., Mayerhofer C., Issels R.D. Differential effects of ifosfamide on dendritic cell-mediated stimulation of T cell interleukin-2 production, natural killer cell cytotoxicity and interferon-gamma production. Clin. Exp. Immunol. 2008; 153: 429–438.
  26. Pukhalsky A., Toptygina A., Khaidukov S. Interleukin-2 receptor β chain as a possible target for low doses of mafosfamide. Med. Inflam. 1995; 4: 175–180.
  27. Pukhalsky A., Shmarina G., Alioshkin V. Cognitive disorders in mice: cytokine signaling pathways as therapeutic targets. OMICS. 2012; 16 (1–2): 71–77.
  28. Matsushita N., Pilon-Thomas S.A., Martin L.M., Riker A.I. Comparative methodologies of regulatory T cell depletion in a murine melanoma model. J. Immunol. Methods. 2008; 333: 167–179.
  29. Radojcic V., Bezak K.B., Skarica M., Pletneva M.A., Yoshimura K., Schulick R.D., Luznik L. Cyclophosphamide resets dendritic cell homeostasis and enhances antitumor immunity through effects that extend beyond regulatory T cell elimination. Cancer Immunol. Immunother. 2010; 59: 137–148.
  30. Stallmach A., Witting B.M., Moser C., Fischinger J., Duchmann R., Zeitz M. Safety and efficacy of intravenous pulse cyclophosphamide in acute steroid refractory inflammatory bowel disease. Gut. 2003; 52: 377–382.
  31. Соколов Е.И., Зыков К.А., Пухальский А.Л., Цыпленкова В.Г., Шевелев В.И. Ингаляция ультрамалых доз алкилирующих препаратов в лечении бронхиальной астмы. Пульмонология. 2002; 12 (3): 82–88.
  32. Prasad S.J., Farrand K.J., Matthews S.A., Chang J.H., McHugh R.S., Ronchese F. Dendritic cells loaded with stressed tumor cells elicit long-lasting protective tumor immunity in mice depleted of CD4+CD25+regulatory T cells. J. Immunol. 2005; 174: 90–98.
  33. Imai H., Saio M., Nonaka K. Depletion of CD4+CD25+ regulatory T cells enhances interleukin-2-induced antitumor immunity in a mouse model of colon adenocarcinoma. Cancer Sci. 2007; 98: 416–423.
  34. Dannull J., Su Z., Rizzieri D., Yang B.K., Coleman D., Yancey D., Zhang A., Dahm P., Chao N., Gilboa E., Vieweg J. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J. Clin. Invest. 2005; 115: 3623–3633.
  35. Salem M.L., Al-Khami A.A., El-Naggar S.A., Díaz-Montero C.M., Chen Y., Cole D.J. Cyclophosphamide induces dynamic alterations in the host microenvironments resulting in a Flt3 ligand-dependent expansion of dendritic cells. J. Immunol. 2010; 184: 1737–1747.
  36. Zarogoulidis P., Papanas N., Kioumis I., Chatzaki E., Maltezos E., Zarogoulidis K. Macrolides: from in vitro anti-inflammatory and immunomodulatory properties to clinical practice in respiratory diseases. Eur. J. Clin. Pharmacol. 2012; 68: 479–503.
  37. Chang D.M., Lan J.L., Lin H.Y., Luo S.F. Dehydroepiandrosterone treatment of women with mild-to-moderate systemic lupus erythematosus: a multicenter randomized, double -blind, placebocontrolled trial. Arthritis Rheum. 2002; 46: 2924–2927.
  38. Petri M.A., Mease P.J., Merrill J.T., Lahita R.G., Iannini M.J., Yocum D.E. et al. Effects of prasterone on disease activity and symptoms in women with active systemic lupus erythematosus . Arthritis Rheum. 2004; 50:2858–2868.
  39. Kasperska-Zajac A., Brzoza Z., Rogala B. Serum concentration of dehydroepiandrosterone sulfate and testosterone in women with severe atopic eczema/dermatitis syndrome. J. Investig. Allergol. Clin. Immunol. 2007; 17: 160–163.
  40. Kannisto S., Laatikainen A., Taivainen A., Savolainen K., Tukiainen H., Voutilainen R. Serum dehydroepiandrosterone sulfate concentration as an indicator of adrenocortical suppression during inhaled steroid therapy in adult asthmatic patients. Eur. J. Endocrinol. 2004; 150: 687–690.
  41. Lin X.H., Choi I.S., Koh Y.A., Cui Y. Effects of combined bacille Calmette -Guerin and dehydroepiandrosterone treatment on established asthma in mice. Exp. Lung Res. 2009; 35: 250–261.
  42. Choi I.S., Cui Y., Koh Y.A., Lee H.C., Cho Y.B., Won Y.H. Effects of dehydroepiandrosterone on Th2 cytokine production in peripheral blood mononuclear cells from asthmatics. Korean J. Intern. Med. 2008; 23: 176–181.
  43. Wenzel S.E., Robinson C.B., Leonard J.M., Panettieri R.A., Jr. Nebulized dehydroepiandrosterone-3-sulfate improves asthma control in the moderate-to-severe asthma results of a 6-week, randomized, double-blind, placebo-controlled study. Allergy Asthma Proc. 2010; 31: 461–471.
  44. Dashtaki R., Whorton A.R., Murphy T.M., Chitano P., Reed W., Kennedy T.P. Dehydroepiandrosterone and analogs inhibit DNA binding of AP-1 and airway smooth muscle proliferation. J. Pharmacol. Exp. Ther. 1998; 285: 876–883.
  45. Koziol-White C.J., Goncharova E.A., Cao G., Johnson M., Krymskaya V.P., Panettieri R.A. Jr. DHEA-S inhibits human neutrophil and human airway smooth muscle migration. Biochеm. Biophys. Acta. 2012; 1822: 1638-1642.
  46. Shah S. European Respiratory Society. 14th Annual Congress. Drug highlights. IDrugs. Glasgow, UK. 2004. P. 914–916.
  47. Wolkowitz O.M., Reus V.I., Roberts E., Manfredi F., Chan T., Raum W.J., Ormiston S., Johnson R., Canick J., Brizendine L., Weingartner H. Dehydroepiandrosterone (DHEA) treatment of depression. Biol. Psychiatry. 1997; 41: 311–318.
  48. Schmidt P.J., Daly R.C., Bloch M., Smith M.J., Danaceau M.A., St Clair L.S., Murphy J.H., Haq N., Rubinow D.R. Dehydroepiandrosterone monotherapy in midlife-onset major and minor depression. Arch. Gen. Psychiatry. 2005; 62: 154–162.
  49. Rabkin J.G., McElhiney M.C., Rabkin R., McGrath P.J. Placebo-controlled trial of dehydroepiandrosterone (DHEA) for treatment of nonmajor depression in patients with HIV/AIDS. Am. J. Psychiatry. 2006; 163: 59–66.
  50. Sergeev G.B., Komarov V.S. The synthesis via criomodification. Mol. Cryst. Liquid Cryst. 2006; 456: 107–115.
  51. Утехина А.Ю., Сергеев Г.Б. Органические наночастицы. Усп. химии. 2011; 80 (3): 233–248.
  52. Nakano D., Hitomi H., Mogi M., Shimada K., Kobori H., Horiuchi M., Sakamoto H., Matsumoto M., Kohno M., Nishiyama A. Blockade of AT1 receptors protects the Entbloodbrain barrier and improves cognition in Dahl salt-sensitive hypertensive rats. Am. J. Hypertens. 2011; 24: 362–368.
  53. Saavedra J.M., Sánchez-Lemus E., Benicky J. Blockade of brain angiotensin II AT1 receptors ameliorates stress, anxiety, brain inflammation and ischemia: Therapeutic implications. Psychoneuroendocrinology. 2011; 36: 1–18.
  54. Saavedra J.M. Angiotensin II AT(1) receptor blockers ameliorate inflammatory stress: a beneficial effect for the treatment of brain disorders. Cell Mol. Neurobiol. 2012; 32: 667–681.

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 1970 "Paediatrician" Publishers LLC



This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies