Methamphetamine administration targets multiple immune subsets and induces phenotypic alterations suggestive of immunosuppression

Robert Harms, Brenda Morsey, Craig W Boyer, Howard S Fox, Nora Sarvetnick, Robert Harms, Brenda Morsey, Craig W Boyer, Howard S Fox, Nora Sarvetnick

Abstract

Methamphetamine (Meth) is a widely abused stimulant and its users are at increased risk for multiple infectious diseases. To determine the impact of meth on the immune system, we utilized a murine model that simulates the process of meth consumption in a typical addict. Our phenotypic analysis of leukocytes from this dose escalation model revealed that meth affected key immune subsets. Meth administration led to a decrease in abundance of natural killer (NK) cells and the remaining NK cells possessed a phenotype suggesting reduced responsiveness. Dendritic cells (DCs) and Gr-1(high) monocytes/macrophages were also decreased in abundance while Gr-1(low) monocytes/macrophages appear to show signs of perturbation. CD4 and CD8 T cell subsets were affected by methamphetamine, both showing a reduction in antigen-experienced subsets. CD4 T cells also exhibited signs of activation, with increased expression of CD150 on CD226-expressing cells and an expansion of KLRG1(+), FoxP3(-) cells. These results exhibit that meth has the ability to disrupt immune homeostasis and impact key subsets of leukocytes which may leave users more vulnerable to pathogens.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Gating strategy for defining major…
Figure 1. Gating strategy for defining major subsets of leukocytes among splenocytes. A.
Dead cells were removed from the analysis using Live/Dead® fixable dead cell stain. B. Doublets were removed from living cells (Live/Dead−) using FSC-A and FSC-H. C. Gr-1bright splenocytes were gated and defined as neutrophils. D. NKp46+ events were gated from Gr-1low/− splenocytes and defined as natural killer (NK) cells. E. NKp46−, Gr-1low/− splenocytes were divided into MHC II+ and MHC II− populations. F. MHC II+ events were split by B220 and CD11c expression. MHC II+, B220+, CD11c− events were defined as B cells. MHC II+ B220−, CD11c+ events were defined as dendritic cells (DCs). G. We split MHC II− events by CD3 and CD11b expression. CD11b-, CD3+, MHC II- events were labeled as T cells. H. CD11b+, CD3−, MHC II- events were split according to SSC profile. Events with higher SSC values (suggesting greater internal granularity) were labeled as eosinophils. I. Events with lower SSC values were divided by Gr-1 expression, giving two populations: Gr-1(Ly-6C)high monocytes/macrophages and Gr-1(Ly-6C)low monocytes/macrophages (mono/MΦ).
Figure 2. Meth causes a reduction of…
Figure 2. Meth causes a reduction of splenic leukocyte subsets.
C57BL/6 mice were treated with vehicle (control) or ramped dosage of methamphetamine (meth – see methods) for 14 days. At day 14, mice were sacrificed and splenic leukocyte subsets were analyzed by flow cytometry. A. Meth treatment did not alter total splenocyte number. B. Meth causes a significant reduction in total natural killer (NK) cells (NKp46+, CD3−, B220−, Gr-1−, MHC II−). C. Gr-1high monocytes (CD11b+, Gr-1high, MHC II−, CD3−, B220−) were reduced after meth treatment. D. Meth treatment reduces dendritic cells (DCs) defined as CD11c+, MHC II+, B220−, Gr-1− events. Dead cells and doublets were removed prior to analysis. **P<0.01 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 3. Meth perturbs Gr-1 low monocytes/macrophages.
Figure 3. Meth perturbs Gr-1low monocytes/macrophages.
Gr-1low monocytes/macrophages were analyzed for signs of activation after meth treatment using flow cytometry. A. Representative histogram showing CD80 (B7-1) expression by Gr-1ow monocytes/macrophages. Red represents a meth-treated animal, black represents a vehicle-treated animal, and grey is the isotype control. B. Meth causes increased expression of CD80 by Gr-1low, quantified by mean fluorescence intensity (MFI). C. Representative histogram showing CD11b (Mac-1) expression by Gr-1low monocytes/macrophages with colors as in A above. D. Meth causes decreased expression of CD11b by Gr-1low monocytes, quantified by MFI. Dead cells and doublets were removed prior to analysis. **p<0.01, ***p<0.001 calculated by Mann-Whitney U Test. Error bars represent SEM. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 4. Meth alters NK cell subsets.
Figure 4. Meth alters NK cell subsets.
We labeled splenic NK cells (NK1.1+, CD4−, CD8−) with antibodies against killer cell lectin-like receptor G1 (KLRG1) and CD27 to determine if meth treatment alters functionally distinct NK cell subsets. A. Representative gating showing KLRG1 and CD27 on gated NK cells. B and C. Meth treatment results in a decreased proportion of KLRG1−, CD27+ NK cells with an increase in proportion of KLRG1+, CD27− NKs. D and E. Although proportionally higher, KLRG1+, CD27− NK cells are not increased in absolute number. However, KLRG1−, CD27+ NK cells were significantly reduced in number. Dead cells and doublets were removed prior to analysis. ***p<0.001 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 5. Meth reduces proportions of CD62L…
Figure 5. Meth reduces proportions of CD62Llow, CD44high splenic CD4 T cells and these cells exhibit lower CD27 expression.
Splenic CD4 T cells were examined after meth treatment to determine if meth alters surface phenotypes suggesting activation/antigen experience and effector status. A. Representative gating showing CD62L and CD44 on CD4 T cells. B. Meth causes a reduction in proportion of CD44high, CD62Llow CD4 T cells. C. CD27 expression by CD44high, CD62Llow CD4 T cells. D. CD44high, CD62Llow CD4 T cells from meth treated animals exhibit a lower proportion of cells expressing CD27 at a high level. **p<0.01, ***p<0.001 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 6. Meth causes reduced proportions of…
Figure 6. Meth causes reduced proportions of alternatively spliced CD4 T cells among the splenic CD62Lhigh compartment.
Meth and control mice were analyzed for surface expression of CD45RB and CD44 on the CD62Lhigh subset. CD45RB expression is downregulated upon antigen experience and can be used to gauge activation levels of CD4 T cells. A. CD45RB and CD44 on CD62Lhigh splenic CD4 T cells. B–D. Meth-treated animals had a higher proportion of CD45RBhigh, CD44low CD4 T cells compared to controls. Meth-treated animals also exhibited lower proportions of CD45RBlow, CD44low and CD45RBlow, CD44highsubsets compared to controls. ***p<0.001 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 7. Meth treatment causes a reduction…
Figure 7. Meth treatment causes a reduction in proportion of CD62Llow, CD44high CD4 T cells in the mesenteric lymph node (MLN).
MLN CD4 T cells were labeled with CD62L and CD44 to determine proportions of antigen-experienced cells. A. CD62L and CD44 on MLN CD4 T cells from meth and vehicle treated animals. B. Meth causes a reduction in proportion of CD44high, CD62Llow CD4 T cells in the MLN. **p<0.01 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 8. Increased expression of CD150 on…
Figure 8. Increased expression of CD150 on CD226+ CD4 T cells after meth treatment.
We examined the expression of the costimulatory markers CD150 and CD226 by splenic CD4 T cells after meth treatment. A. Representative gating and histogram showing selection of CD226+ CD4 T cells and increased expression of CD150 by meth treated animals compared to control. Red represents a meth-treated animal, black is vehicle-treated animal, and grey is the isotype control. B. CD226+ CD4 T cells from meth treated animals express higher levels of CD150 than those from control animals. Protein expression was calculated using mean fluorescence intensity (MFI). ***p<0.001 calculated by Mann-Whitney U Test. Error bars represent SEM. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 9. Meth promotes the expansion of…
Figure 9. Meth promotes the expansion of splenic KLRG1+, FoxP3− CD4 T cells.
Meth-treated animals possessed an expansion of KLRG1+ CD4 T cells within the spleen. These cells were CD44high and negative/low for FoxP3, CD25, CD62L, NKG2D, and CD45RB. A. Representative gating showing significant expansion of splenic KLRG1+, FoxP3− CD4 T cells among the CD44high, CD45RBlow compartment with relatively unchanged proportions of FoxP3-expressing subsets after meth treatment. B and C. Proportion and absolute number of KLRG1+, FoxP3− CD4 cells are increased with meth. ***p<0.001 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 10. Meth-treated animals exhibit a greater…
Figure 10. Meth-treated animals exhibit a greater proportion of naïve phenotype CD8 T cells and reduced proportions of antigen-experienced subsets within the spleen.
Splenic CD8 T cells were labeled with CD62L and CD44 and classified into 4 groups according the expression of these two proteins: CD62Lhigh, CD44low (Naïve CD8 T cell, TN), CD62Lhigh, CD44high, (central memory CD8 T cell, TCM), CD62Llow, CD44high (effector memory CD8 T cells, TEM), and CD62Llow, CD44low (acute effector CD8 T cell, TAE). A. Gating showing CD44 and CD62L expression by splenic CD8 T cells. B–E. After meth treatment, the TN compartment is increased in proportion, while the TAE and TEM showed a decrease in proportion and TCM appeared unchanged. *p<0.05, **p<0.01 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 11. CD8 T cells from the…
Figure 11. CD8 T cells from the mesenteric lymph node (MLN) also show reductions in proportion of antigen-experienced subsets with an increase in the naïve compartment.
Mesenteric lymph node CD8 T cells were labeled with CD62L and CD44 and classified as in Fig. 10. A. Gating showing CD44 and CD62L expression by MLN CD8 T cells. B–E. After meth treatment, the TN compartment is increased in proportion, while the TAE, TEM, and TCM compartments all revealed a decrease in proportion. **p<0.01, ***p<0.001 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 12. Meth causes a reduced proportion…
Figure 12. Meth causes a reduced proportion and number of KLGR1+ CD8 T cells in the MLN.
CD8 T cells were labeled with KLRG1 to investigate whether meth impacts subsets of antigen-experienced CD8 T cells. A. The proportion and number of CD8 T cells expressing KLRG1 in the MLN is reduced after meth treatment. *p<0.01, ***p<0.001 calculated by Mann-Whitney U Test. Bars represent mean. Data are from 2 experiments of 5 animals per treatment group per experiment.
Figure 13. Methamphetamine impacts the innate and…
Figure 13. Methamphetamine impacts the innate and adaptive arms of immunity.
Summary of the effects of methamphetamine on immune cell subsets based upon the observations in this report.

References

    1. Brouwer KC, Case P, Ramos R, Magis-Rodriguez C, Bucardo J, et al. (2006) Trends in production, trafficking, and consumption of methamphetamine and cocaine in Mexico. Substance Use and Misuse 41: 707–727.
    1. Cronkwright Kirkos W, Carrique T, Griffen K, La Barge AP (2008) The York region methamphetamine strategy. Canadian Medical Association Journal 178: 1655–1656.
    1. Degenhardt L, Baker A, Maher L (2008) Methamphetamine: Geographic areas and populations at risk, and emerging evidence for effective interventions. Drug and Alcohol Review 27: 217–219.
    1. McKetin R, Kozel N, Douglas J, Ali R, Vicknasingam B, et al. (2008) The rise of methamphetamine in southeast and east Asia. Drug and Alcohol Review 27: 220–228.
    1. Mau MK, Asao K, Efird J, Saito E, Ratner R, et al. (2009) Risk factors associated with methamphetamine use and heart failure among native Hawaiians and other pacific island peoples. Journal of Vascular Health and Risk Management 5: 45–52.
    1. Sheridan J, Butler R, Wheeler A (2009) Initiation into methamphetamine use: qualitative findings from an exploration of first time use among a group of New Zealand users. Journal of Psychoactive Drugs 41: 11–17.
    1. Bonell CP, Hickson FC, Weatherburn P, Reid DS (2010) Methamphetamine use among gay men across the UK. The International Journal on Drug Policy 21: 244–246.
    1. Gonzales R, Mooney L, Rawson RA (2010) The methamphetamine problem in the United States. Annual Review of Public Health 31: 385–398.
    1. Peltzer K, Ramlagan S, Johnson BD, Phaswana-Mafuya N (2010) Illicit drug use and treatment in South Africa: a review. Substance Use and Misuse 45: 2221–2243.
    1. Nicosia N, Pacula RL, Kilmer B, Lundberg R, Chiesa J (2009) The economic cost of methamphetamine use in the United States, 2005. Santa Monica, CA: RAND Corporation. Available: . Accessed 2010 June 22.
    1. Cruickshank CC, Dyer KR (2009) A review of the clinical pharmacology of methamphetamine. Addiction 104: 1085–1099.
    1. Rivière GJ, Gentry WB, Owens SM (2000) Disposition of methamphetamine and its metabolite amphetamine in brain and other tissues in rats after intravenous administration. The Journal of Pharmacology and Experimental Therapeutics 292: 1042–1047.
    1. Volkow ND, Fowler JS, Wang GJ, Shumay E, Telang F, et al. (2010) Distribution and pharmacokinetics of methamphetamine in the human body: clinical implications. PLoS ONE 5: e15269.
    1. Nordahl TE, Salo R, Leamon M (2003) Neuropsychological effects of chronic methamphetamine use on neurotransmitters and cognition: a review. Journal of Neuropsychiatry and Clinical Neuroscience 15: 317–325.
    1. Krasnova IN, Cadet JL (2009) Methamphetamine toxicity and messengers of death. Brain Research Reviews 60: 379–407.
    1. Hutin YJF, Sabin KM, Hutwagner LC, Schaben L, Shipp GM, et al. (2000) Multiple modes of hepatitis A virus transmission among methamphetamine users. American Journal of Epidemiology 152: 186–192.
    1. Gonzales RP, Marinelli-Casey P, Shoptaw S, Ang A, Rawson RA (2006) Hepatitis C virus infection among methamphetamine-dependent individuals in outpatient treatment. Journal of Substance Abuse Treatment 31: 195–202.
    1. Vogt TM, Perz JF, Van Houten Jr CK, Harrington R, Hansuld T, et al. (2006) An outbreak of hepatitis B virus among methamphetamine injectors: the role of sharing injection drug equipment. Addiction 101: 726–730.
    1. Cohen AL, Shuler C, McAllister S, Fosheim GE, Brown MG, et al. (2007) Methamphetamine use and methicillin-resistant Staphylococcus aureus skin infections. Emerging Infectious Diseases 13: 1707–1713.
    1. Corsi KF, Booth RE (2008) HIV sex risk behaviors among heterosexual methamphetamine users: literature review from 2000 to present. Current Drug Abuse Reviews 1: 292–296.
    1. Forrest DW, Metsch LR, LaLota M, Cardenas G, Beck DW, et al. (2010) Crystal methamphetamine use and sexual risk behaviors among HIV-positive and HIV-negative men who have sex with men in south Florida. Journal of Urban Health: Bulleting of the New York Academy of Medicine 87: 480–485.
    1. Freeman P, Walker BC, Harris DR, Garofalo R, Willard N, et al. (2011) Methamphetamine use and risk for HIV among young men who have sex with men in 8 US cities. Archives of Pediatric and Adolescent Medicine 165: 736–740.
    1. Marshall BDL, Wood E, Shoveller JA, Patterson TL, Montaner JSG, et al. (2011) Pathways to HIV risk and vulnerability among lesbian, gay, bisexual, and transgendered methamphetamine users: A multi-cohort gender-based analysis. BMC Public Health 11: 20.
    1. Liang H, Wang X, Chen H, Song L, Ye L, et al. (2008) Methamphetamine enhances HIV infection of macrophages. American Journal of Pathology 172: 1617–1624.
    1. Ye L, Peng JS, Wang X, Wang YJ, Luo GX, et al. (2008) Methamphetamine enhances hepatitis C virus replication in human hepatocytes. Journal of Viral Hepatology 15: 261–270.
    1. Gagnon L, Lacroix F, Chan J, Buttar HS (1992) In vitro effects of ‘designer’ amphetamines on human peripheral blood mononuclear leukocytes proliferation and on natural killer cell activity. Toxicology Letters 63: 313–319.
    1. Yu Q, Zhang D, Walston M, Zhang J, Liu Y, et al. (2002) Chronic methamphetamine exposure alters immune function in normal and retrovirus-infected mice. International Immunopharmacology 2: 951–962.
    1. Potula R, Hawkins BJ, Cenna JM, Fan S, Dykstra H, et al. (2010) Methamphetamine causes mitrochondrial oxidative damage in human T lymphocytes leading to functional impairment. Journal of Immunology 185: 2867–2876.
    1. House RV, Thomas PT (1994) Comparison of immune functional parameters following in vitro exposure to natural and synthetic amphetamines. Immunopharmacology and Immunotoxicology 16: 1–21.
    1. In S, Son E, Rhee D, Pyo S (2005) Methamphetamine administration produces immunomodulation in mice. Journal of Toxicology and Environmental Health, Part A 68: 2133–2145.
    1. Wey S, Wu H, Chang F, Jan T (2008) Methamphetamine and diazepam suppress antigen-specific cytokine expression and antibody production in ovalbumin-sensitized BALB/c mice. Toxicology Letters 181: 157–162.
    1. In S, Son E, Rhee D, Pyo S (2004) Modulation of murine macrophage function by methamphetamine. Journal of Toxicology and Environmental Health, Part A 67: 1923–1937.
    1. Tallóczy Z, Martinez J, Joset D, Ray Y, Gácser A, et al. (2008) Methamphetamine inhibits antigen processing, presentation, and phagocytosis. PLoS Pathogens 4: e28.
    1. Martinez LR, Mihu MR, Gácser A, Santambrogio L, Nosanchuk JD (2009) Methamphetamine enhances histoplasmosis by immunosupression of the host. The Journal of Infectious Diseases 200: 131–141.
    1. Marcondes MCG, Flynn C, Watry DD, Zandonatti M, Fox HS (2010) Methamphetamine increases brain viral load and activates natural killer cells in simian immunodeficiency virus-infected monkeys. The American Journal of Pathology 177: 355–361.
    1. Saito M, Yamaguchi T, Kawata T, Ito H, Kanai T, et al. (2005) Effects of methamphetamine on cortisone concentration, NK cell activity and mitogen response of T-lymphocytes in female cynomolgus monkeys. Experimental Animals 55: 477–481.
    1. Saito M, Terada M, Kawata T, Ito H, Shigematsu N, et al. (2008) Effects of single or repeated administrations of methamphetamine on immune response in mice. Experimental Animals 57: 35–43.
    1. Thomas DM, Walker PD, Benjamins JA, Geddes TJ, Kuhn DM (2004) Methamphetamine neurotoxicity in dopamine nerve endings of the striatum is associated with microglia activation. The Journal of Pharmacology and Experimental Therapeutics 311: 1–7.
    1. Iwasa M, Maeno Y, Inoue H, Koyama H, Matoba R (1996) Induction of apoptotic cell death in rat thymus and spleen after bolus injection of methamphetamine. International Journal of Legal Medicine 109: 23–28.
    1. Daley JM, Thomay AA, Connolly MD, Reichner JS, Albina JE (2008) Use of Ly6G-specific monoclonal antibody to deplete neutrophils in mice. Journal of Leukocyte Biology 83: 64–70.
    1. Strauss-Ayali D, Conrad SM, Mosser DM (2007) Monocyte subpopulations and their differentiation patterns during infection. Journal of Leukocyte Biology 82: 244–252.
    1. Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, et al. (2009) Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 325: 612–616.
    1. Walzer T, Bléry M, Chaix J, Fuseri N, Chasson L, et al. (2007) Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46. Proceedings of the National Academy of Sciences 104: 3384–3389.
    1. Sharpe AH, Freeman GJ (2002) The B7-CD28 superfamily. Nature Reviews Immunology 2: 116–126.
    1. Hayakawa Y, Smyth MJ (2006) CD27 dissects mature NK cells into two subsets with distinct responsiveness and migratory capacity. Journal of Immunology 176: 1517–1524.
    1. Huntington ND, Tabarias H, Fairfax K, Brady J, Hayakawa Y, et al. (2007) NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation. Journal of Immunology 178: 4764–4770.
    1. Howie D, Okamoto S, Rietdijk S, Clarke K, Wang N, et al. (2002) The role of SAP in murine CD150 (SLAM) -mediated T-cell proliferation and interferon γ production. Blood 100: 2899–2907.
    1. Dardalhon V, Schubart AS, Reddy J, Meyers JH, Monney L, et al. (2005) CD226 is specifically expressed on the surface of Th1 cells and regulates their expansion and effector functions. Journal of Immunology 175: 1558–1565.
    1. Wing K, Sakaguchi S (2010) Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nature Immunology 11: 7–13.
    1. Beyersdorf N, Ding X, Tietze JK, Hanke T (2007) Characterization of mouse CD4 T cell subsets defined by expression of KLRG1. European Journal of Immunology 37: 3445–3454.
    1. Tsujimura K, Obata Y, Matsudaira Y, Nishida K, Akatsuka Y, et al. (2006) Characterization of murine CD160+ CD8+ T lymphocytes. Immunology Letters 106: 48–56.
    1. Wiede F, Roomberg A, Cretney E, Lechner A, Fromm P, et al. (2009) Age-dependent, polyclonal hyperactivation of T cells is reduced in TNF-negative gld/gld mice. The Journal of Leukocyte Biology 85: 108–116.
    1. Sarkar S, Kalia V, Haining WN, Konieczny BT, Subramaniam S, et al. (2008) Functional and genomic profiling of effector CD8 T cell subsets with distinct memory fates. The Journal of Experimental Medicine 205: 625–640.
    1. Kojima T, Okamoto I, Miyazaki T, Chikasue F, Yashiki M, et al. (1986) Detection of methamphetamine and amphetamine in a skeletonized body buried for 5 years. Forensic Science International 31: 93–102.
    1. Nagata T, Kimura K, Hara K, Kudo K (1990) Methamphetamine and amphetamine concentrations in postmortem rabbit tissues. Forensic Science International 48: 39–47.
    1. Sato Y, Kondo K, Takayasu T, Ohshima T (2000) Detection of methamphetamine in a severely burned cadaver – a case report. Nihon Hoigaku Zasshi 54: 420–424.
    1. Geissmann F, Jung S, Littman DR (2003) Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19: 71–82.
    1. Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, et al. (2007) The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. The Journal of Experimental Medicine 204: 3037–3047.
    1. Tacke F, Ginhoux F, Jakubzick C, van Rooijen N, Merad M, et al. (2006) Immature monocytes acquire antigens from other cells in the bone marrow and present them to T cells after maturing in the periphery. The Journal of Experimental Medicine 203: 583–597.
    1. Peng Y, Latchman Y, Elkon KB (2009) Ly6Clow monocytes differentiate into dendritic cells and cross-tolerize T cells through PDL-1. Journal of Immunology 182: 2777–2785.
    1. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ (2007) Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity 27: 111–122.
    1. Butte MJ, Peña-Cruz V, Kim MJ, Freeman GJ, Sharpe AH (2008) Interaction of human PD-L1 and B7–1. Molecular Immunology 45: 3567–3572.
    1. Park JJ, Omiya R, Matsumura Y, Sakoda Y, Kuramasu A, et al. (2010) B7-H1/CD80 interaction is required for the induction and maintenance of peripheral T-cell tolerance. Blood 116: 1291–1298.
    1. Bromley SK, Iaboni A, Davis SJ, Whitty A, Green JM, et al. (2001) The immunological synapse and CD28-CD80 interactions. Nature Immunology 2: 1159–1166.
    1. Schif-Zuck S, Gross N, Assi S, Rostoker R, Serhan CN, et al. (2011) Saturated-efferocytosis generates pro-resolving CD11blow macrophages: modulation by resolvins and glucocorticoids. European Journal of Immunology 41: 366–379.
    1. Marcenaro E, Carlomagno S, Pesce S, Della Chiesa M, Parolini S, et al. (2011) NK cells and their receptors during viral infections. Immunotherapy 3: 1075–1086.
    1. Waldhauer I, Steinle A (2008) NK cells and cancer immunosurveillance. Oncogene 27: 2932–5943.
    1. Waggoner SN, Cornberg M, Selin LK, Welsh RM (2011) Natural killer cells act as rheostats modulating antiviral T cells. Nature 481: 394–398.
    1. Chiossone L, Chaix J, Fuseri N, Roth C, Vivier E, et al. (2009) Maturation of mouse NK cells is a 4-stage developmental program. Blood 113: 5488–5496.
    1. Robbins SH, Nguyen KB, Takahashi N, Mikayama T, Biron CA, et al. (2002) Cutting edge: Inhibitory functions of the killer cell lectin-like receptor G1 molecule during the activation of mouse NK cells. Journal of Immunology 168: 2585–2589.
    1. Denoeud J, Moser M (2011) Role of CD27/CD70 pathway of activation in immunity and tolerance. Journal of Leukocyte Biology 89: 195–203.
    1. Hermiston ML, Xu Z, Weiss A (2003) CD45: A critical regulator of signaling thresholds in immune cells. Annual Review of Immunology 21: 107–137.
    1. Ellis RJ, Childers ME, Cherner M, Lazzaretto D, Letendre S, et al. (2003) Increased human immunodeficiency virus loads in active methamphetamine users are explained by reduced effectiveness of antiretroviral therapy. The Journal of Infectious Diseases 188: 1820–1826.
    1. Henning G, Kraft MS, Derfuss T, Pirzer R, de Saint-Basile G, at al (2001) Signaling lymphocyte activation molecule (SLAM) regulates T cellular cytotoxicity. European Journal of Immunology 31: 2741–2750.
    1. Ito M, Maruyama T, Saito N, Koganei S, Yamamoto K, et al. (2006) Killer cell lectin-like receptor G1 binds three members of the classical cadherin family to inhibit NK cell cytotoxicity. The Journal of Experimental Medicine 203: 289–295.
    1. Li Y, Hofmann M, Wang Q, Teng L, Chlewicki LK, et al. (2009) Structure of natural killer cell receptor KLRG1 bound to E-cadherin reveals basis for MHC-independent missing self recognition. Immunity 31: 35–46.
    1. Reiley WW, Shafiani S, Wittmer ST, Tucker-Heard G, Moon JJ, et al. (2010) Distinct functions of antigen-specific CD4 T cells during Mycobacterium tuberculosis infection. Proceedings of the National Academy of Sciences 107: 19408–19413.

Source: PubMed

3
Sottoscrivi