Systemic DPP4 activity is reduced during primary HIV-1 infection and is associated with intestinal RORC+ CD4+ cell levels: a surrogate marker candidate of HIV-induced intestinal damage

Mickaël J Ploquin, Armanda Casrouge, Yoann Madec, Nicolas Noël, Beatrice Jacquelin, Nicolas Huot, Darragh Duffy, Simon P Jochems, Luca Micci, Camille Lécuroux, Faroudy Boufassa, Thijs Booiman, Thalia Garcia-Tellez, Mathilde Ghislain, Roger Le Grand, Olivier Lambotte, Neeltje Kootstra, Laurence Meyer, Cecile Goujard, Mirko Paiardini, Matthew L Albert, Michaela Müller-Trutwin, Mickaël J Ploquin, Armanda Casrouge, Yoann Madec, Nicolas Noël, Beatrice Jacquelin, Nicolas Huot, Darragh Duffy, Simon P Jochems, Luca Micci, Camille Lécuroux, Faroudy Boufassa, Thijs Booiman, Thalia Garcia-Tellez, Mathilde Ghislain, Roger Le Grand, Olivier Lambotte, Neeltje Kootstra, Laurence Meyer, Cecile Goujard, Mirko Paiardini, Matthew L Albert, Michaela Müller-Trutwin

Abstract

Introduction: Combined anti-retroviral therapy (cART) transformed HIV-1 from a deadly disease into a chronic infection, but does not cure HIV infection. It also does not fully restore HIV-induced gut damage unless administered extremely early after infection. Additional biomarkers are needed to evaluate the capacity of therapies aimed at HIV remission/cure to restore HIV-induced intestinal immune damage and limit chronic inflammation. Herein, we aimed to identify a systemic surrogate marker whose levels would reflect gut immune damage such as intestinal Th17 cell loss starting from primary HIV-1 infection.

Methods: Biomarker discovery approaches were performed in four independent cohorts, covering HIV-1 primary and chronic infection in 496 naïve or cART-treated patients (Amsterdam cohort (ACS), ANRS PRIMO, COPANA and CODEX cohorts). The concentration and activity of soluble Dipeptidylpeptidase 4 (sDPP4) were quantified in the blood from these patients, including pre- and post-infection samples in the ACS cohort. For quantification of DPP4 in the gut, we utilized two non-human primate models, representing pathogenic (macaque) and non-pathogenic (African green monkey) SIV infection. Four gut compartments were analysed in each animal model (ileum, jejunum, colon and rectum) for quantification of DPP4, RORC and TBX21 gene expression in sorted CD4+ cells. To analyse if sDPP4 levels increase when Th17 cells were restored, we quantified sDPP4 in plasma from SIV-infected macaques treated with IL-21.

Results: We showed that sDPP4 levels were strongly decreased in primary HIV-1 infection. Strikingly, sDPP4 levels in primary HIV-1 infection predicted time to AIDS. They were not increased by cART in chronic HIV-1 infection (median 36 months on cART). In the gut of SIV-infected non-human primates, DPP4 mRNA was higher in CD4+ than CD4- leucocytes. DPP4 specifically correlated with RORC expression, a Th17 marker, in CD4+ cells from the intestine. We further demonstrated that sDPP4 activity levels were increased in animals treated with IL-21 and that this increase was associated with restoration of the Th17 compartment and reduced inflammation. Furthermore, DPP4 mRNA levels in small intestine CD4+ cells positively correlated with circulating DPP4 activity.

Conclusion: These data provide evidence that blood sDPP4 levels could be useful as a correlate for HIV-induced intestinal damage.

Keywords: HIV; SIV; Th17; biomarker; dipeptidylpeptidase; inflammation; intestine.

© 2018 The Authors. Journal of the International AIDS Society published by John Wiley & sons Ltd on behalf of the International AIDS Society.

Figures

Figure 1
Figure 1
Soluble DPP4 levels in blood over time during distinct phases of HIV‐1 infection. (a) Soluble DPP4 activity levels over time in patients from the Amsterdam cohort Study (ACS). Blood collected before HIV‐1 infection and at early time points post‐infection were analysed. (b). Soluble DPP4 activity levels in 11 of these patients from the ACS for whom samples at all four time points of the study were available. (c) Soluble DPP4 activity levels in healthy donors (HD), in HIV‐infected treatment‐naive patients at early time points of infection (primary infection, PHI and six months post‐PHI) from the ANRS CO6 cohort, in the chronic phase of infection (viremic patients (VIR)) from the ANRS COPANA cohort, and during controlled infection, either cART‐treated patients (cART) from the COPANA cohort and HIV controllers (HIC) from the ANRS CODEX cohort. (d‐f) Evolution of sDPP4 levels before and during cART in 40 patients from the ANRS COPANA cohort. The values from the same patient are connected through a line to show the individual evolution of the sDPP4 levels before and after cART initiation. (d) Pre‐ and post‐ART sDPP4 activity (UI/mL) in the 40 cART‐treated patients. Twenty of these patients had received Protease inhibitors (PI). (e) Pre‐ and post‐ART sDPP4 activity (UI/mL) in 20 NRTI‐based cART‐treated patients. (f) Pre‐ and post‐ART sDPP4 activity (UI/mL) in the 20 patients on cART with PI‐containing regimen. Pre‐inf. = before HIV‐1 infection; PHI =  primary HIV‐1 infection; M3 = three months post‐seroconversion; M6 = 6 months post‐seroconversion (panel a‐b in the ACS) or post‐PHI (panel C in the PRIMO cohort). For graphs a, b and c, the median +interquartile range are shown. For graphs a and c, the student t‐test was used. For graphs b, d, e and f, the Wilcoxon sign‐rank test for paired data was used. *< 0.05; **< 0.01; ***< 0.001; ****< 0.0001.
Figure 2
Figure 2
Soluble blood DPP4 levels with regard to disease progression profiles. Soluble DPP4 levels were quantified during primary HIV‐1 infection in patients from the ANRS PRIMO cohort displaying distinct profiles of disease progression (SP, slow progressors; P, normal progressors; RP, rapid progressors; HD, healthy donors). (a) sDPP4 enzymatic activity per ml of blood. The median levels were 18.0 for HD, 4.4 for SP and P and 3.8 for RP. (b) Kaplan–Meier survival analysis of AIDS‐related death by sDPP4 levels measured six months after seroconversion (M6) (≤ or > to the median) in treatment‐naïve patients from the Amsterdam cohort. The dotted line corresponds to sDPP4 levels > median and the solid line to sDDP4 levels < median at M6. For graph a, the Wilcoxon non‐parametric test was used and for graph b the log‐rank test. The median and interquartile range is shown in graph a. **< 0.01; ***< 0.001; ****< 0.0001.
Figure 3
Figure 3
DPP4 mRNA levels in the gut in pathogenic and non‐pathogenic SIV infection. (a,c,e) Four intestinal compartments (ileum, jejunum, colon, rectum) from five rhesus macaques and five AGM were studied at an early phase of infection (day 65 pi.). CD4+ leucocytes were enriched from the distinct sections of the intestine. The values for the small intestine (ileum, jejunum) and large intestine (rectum, colon) were pooled. For MAC, the available material was: jejunum, colon and rectum for five animals, ileum for three to five animals; For AGM, the available samples were: jejunum, colon and rectum for four to five animals, ileum for four to five animals). When the material was limited, we privileged the analyses of DPP4mRNA. (a) DPP4 mRNA levels in intestinal CD4+ cells. (b) DPP4 mRNA expression in CD4+ cells from the small intestine (jejunum) from the five MAC and five AGM plotted against plasma sDPP4 activity in blood from the same animals. (c) RORCmRNA levels in intestinal CD4+ cells. (d) RORCmRNA expression levels plotted against DPP4 mRNA in CD4+ cells from the small intestine. (e) TBX21 (t‐bet) mRNA levels in intestinal CD4+ cells. (f) TBX21 mRNA expression levels plotted against DPP4 mRNA in CD4+ cells from the small intestine. The correlations were statistically significant when the two species were pooled, but not within individual species. (b,d,f) Each circle represents the value of one tissue sample. Black (full circles): MAC; white (open circles): AGM. For graphs a, c and e the Wilcoxon non‐parametric test was used; for graphs b, d and f the Spearman non‐parametric correlation was used. The median and interquartile range are shown in graphs a, c and e. FC, fold change. *<  0.05; **< 0.01; ***< 0.001.
Figure 4
Figure 4
sDPP4 dynamics in blood after IL‐21 immunotherapy of SIV‐infected macaques. (a) sDPP4 activity in blood was measured in a longitudinal analysis before initiation of IL‐21 therapy (week 2 p.i.), at the end of IL‐21 treatment (week 6 p.i.) and after IL‐21 treatment (Weeks 10, 18 and 23 p.i.). The ratio of pre‐ and post‐treatment sDPP4 levels are shown. (b and c) IL‐17+ and IL‐17+ IFN‐•+ cells were measured in rectal biopsies on week 4 and week 6. (b) Correlation between the percentage of intestinal IL‐17+ IFN‐•+ cells and the plasma sDPP4 activity (fold change from week 2 to week 10) (c) Correlation between the percentage of intestinal IL‐17+ cells and plasma sDPP4 activity (fold change from week 2 to week 10). (d‐f) Correlation between sDPP4 activity in blood (fold change between week 2 and 10) and levels of (d) Ki67+ CD4+ T cells in gut (e) blood IP‐10 levels (f) blood sCD14 and (g) blood LPS after IL‐21 therapy cessation at week 23 p.i. For graph a, the Wilcoxon non‐parametric test was used; for graphs b to g, the Spearman non‐parametric correlation. The median and interquartile range are shown in graph a. *< 0.05; **< 0.01. Six rhesus macaques infected with SIVmac and treated with IL‐21 and six rhesus control macaques infected with SIVmac were analysed. CTRL: control monkeys; +IL‐21: IL‐21‐treated monkeys.

References

    1. Maartens G, Celum C, Lewin SR. HIV infection: epidemiology, pathogenesis, treatment, and prevention. Lancet. 2014;384:258–71.
    1. Barré‐Sinoussi F, Ross AL, Delfraissy J‐F. Past, present and future: 30 years of HIV research. Nat Rev Microbiol. 2013;11:877–83.
    1. Katlama C, Deeks SG, Autran B, Martinez‐Picado J, van Lunzen J, Rouzioux C, et al. Barriers to a cure for HIV: new ways to target and eradicate HIV‐1 reservoirs. Lancet. 2013;381:2109–17.
    1. Churchill MJ, Deeks SG, Margolis DM, Siliciano RF, Swanstrom R. HIV reservoirs: what, where and how to target them. Nat Rev Microbiol. 2016;14:55–60.
    1. Deleage C, Wietgrefe SW, Del Prete G, Morcock DR, Hao XP, Piatak M, et al. Defining HIV and SIV reservoirs in lymphoid tissues. Pathog Immun. 2016;1:68–106.
    1. Sereti I, Krebs SJ, Phanuphak N, Fletcher JL, Slike B, Pinyakorn S, et al. Persistent, albeit reduced, chronic inflammation in persons starting antiretroviral therapy in acute HIV infection. Clin Infect Dis. 2017;64:124–31.
    1. Boulware DR, Hullsiek KH, Puronen CE, Rupert A, Baker JV, French MA, et al. Higher levels of CRP, D‐dimer, IL‐6, and hyaluronic acid before initiation of antiretroviral therapy (ART) are associated with increased risk of AIDS or death. J Infect Dis. 2011;203:1637–46.
    1. French MA, King MS, Tschampa JM, da Silva BA, Landay AL. Serum immune activation markers are persistently increased in patients with HIV infection after 6 years of antiretroviral therapy despite suppression of viral replication and reconstitution of CD4+ T cells. J Infect Dis. 2009;200:1212–5.
    1. Hunt PW, Brenchley J, Sinclair E, McCune JM, Roland M, Page‐Shafer K, et al. Relationship between T cell activation and CD4+ T cell count in HIV‐seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J Infect Dis. 2008;197:126–33.
    1. Noel N, Boufassa F, Lécuroux C, Saez‐Cirion A, Bourgeois C, Dunyach‐Remy C, et al. Elevated IP10 levels are associated with immune activation and low CD4+ T‐cell counts in HIV controller patients. AIDS. 2014;28:467–76.
    1. Krishnan S, Wilson EMP, Sheikh V, Rupert A, Mendoza D, Yang J, et al. Evidence for innate immune system activation in HIV type 1‐infected elite controllers. J Infect Dis. 2014;209:931–9.
    1. Borges ÁH, Silverberg MJ, Wentworth D, Grulich AE, Fätkenheuer G, Mitsuyasu R, et al. Predicting risk of cancer during HIV infection: the role of inflammatory and coagulation biomarkers. AIDS. 2013;27:1433–41.
    1. Nordell AD, McKenna M, Borges ÁH, Duprez D, Neuhaus J, Neaton JD, et al. Severity of cardiovascular disease outcomes among patients with HIV is related to markers of inflammation and coagulation. J Am Heart Assoc. 2014;3:e000844.
    1. Siliciano JD, Siliciano RF. Recent developments in the effort to cure HIV infection: going beyond N = 1. J Clin Invest. 2016;126:409–14.
    1. Lewin SR, Deeks SG, Barré‐Sinoussi F. Towards a cure for HIV–are we making progress? Lancet. 2014;384:209–11.
    1. Paiardini M, Müller‐Trutwin M. HIV‐associated chronic immune activation. Immunol Rev. 2013;254:78–101.
    1. Zevin AS, McKinnon L, Burgener A, Klatt NR. Microbial translocation and microbiome dysbiosis in HIV‐associated immune activation. Curr Opin HIV AIDS. 2016;11:182–90.
    1. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–71.
    1. Schuetz A, Deleage C, Sereti I, Rerknimitr R, Phanuphak N, Phuang‐Ngern Y, et al. Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV‐related immune activation. PLoS Pathog. 2014;10:e1004543.
    1. Deleage C, Schuetz A, Alvord WG, Johnston L, Hao X‐P, Morcock DR, et al. Impact of early cART in the gut during acute HIV infection. JCI Insight. 2016;1:e87065.
    1. Mudd JC, Brenchley JM. Gut mucosal barrier dysfunction, microbial dysbiosis, and their Role in HIV‐1 disease progression. J Infect Dis. 2016;214(Suppl 2):S58–66.
    1. Somsouk M, Dunham RM, Cohen M, Albright R, Abdel‐Mohsen M, Liegler T, et al. The immunologic effects of mesalamine in treated HIV‐infected individuals with incomplete CD4+ T cell recovery: a randomized crossover trial. PLoS ONE. 2014;9:e116306.
    1. Kristoff J, Haret‐Richter G, Ma D, Ribeiro RM, Xu C, Cornell E, et al. Early microbial translocation blockade reduces SIV‐mediated inflammation and viral replication. J Clin Invest. 2014;124:2802–6.
    1. Sandler NG, Zhang X, Bosch RJ, Funderburg NT, Choi AI, Robinson JK, et al. Sevelamer does not decrease lipopolysaccharide or soluble CD14 levels but decreases soluble tissue factor, low‐density lipoprotein (LDL) cholesterol, and oxidized LDL cholesterol levels in individuals with untreated HIV infection. J Infect Dis. 2014;210:1549–54.
    1. Tenorio AR, Chan ES, Bosch RJ, Macatangay BJC, Read SW, Yesmin S, et al. Rifaximin has a marginal impact on microbial translocation, T‐cell activation and inflammation in HIV‐positive immune non‐responders to antiretroviral therapy ‐ ACTG A5286. J Infect Dis. 2015;211:780–90.
    1. Manuzak JA, Hensley‐McBain T, Zevin AS, Miller C, Cubas R, Agricola B, et al. Enhancement of microbiota in healthy macaques results in beneficial modulation of mucosal and systemic immune function. J Immunol. 2016;196:2401–9.
    1. Hensley‐McBain T, Zevin AS, Manuzak J, Smith E, Gile J, Miller C, et al. Effects of fecal microbial transplantation on microbiome and immunity in simian immunodeficiency virus‐infected macaques. J Virol. 2016;90:4981–9.
    1. Ortiz AM, Klase ZA, DiNapoli SR, Vujkovic‐Cvijin I, Carmack K, Perkins MR, et al. IL‐21 and probiotic therapy improve Th17 frequencies, microbial translocation, and microbiome in ARV‐treated, SIV‐infected macaques. Mucosal Immunol. 2016;9:458–67.
    1. d'Ettorre G, Ceccarelli G, Giustini N, Serafino S, Calantone N, De Girolamo G, et al. Probiotics reduce inflammation in antiretroviral treated, HIV‐infected individuals: results of the “Probio‐HIV” clinical trial. PLoS ONE 2015;10:e0137200.
    1. Klatt NR, Canary LA, Sun X, Vinton CL, Funderburg NT, Morcock DR, et al. Probiotic/prebiotic supplementation of antiretrovirals improves gastrointestinal immunity in SIV‐infected macaques. J Clin Invest. 2013;123:903–7.
    1. Pallikkuth S, Micci L, Ende ZS, Iriele RI, Cervasi B, Lawson B, et al. Maintenance of intestinal Th17 cells and reduced microbial translocation in SIV‐infected rhesus macaques treated with interleukin (IL)‐21. PLoS Pathog. 2013;9:e1003471.
    1. Chevalier MF, Petitjean G, Dunyach‐Rémy C, Didier C, Girard P‐M, Manea ME, et al. The Th17/Treg ratio, IL‐1RA and sCD14 levels in primary HIV infection predict the T‐cell activation set point in the absence of systemic microbial translocation. PLoS Pathog. 2013;9:e1003453.
    1. Marchetti G, Cozzi‐Lepri A, Merlini E, Bellistrì GM, Castagna A, Galli M, et al. Microbial translocation predicts disease progression of HIV‐infected antiretroviral‐naive patients with high CD4+ cell count. AIDS. 2011;25:1385–94.
    1. León A, Leal L, Torres B, Lucero C, Inciarte A, Arnedo M, et al. Association of microbial translocation biomarkers with clinical outcome in controllers HIV‐infected patients. AIDS. 2015;29:675–81.
    1. Abad‐Fernández M, Vallejo A, Hernández‐Novoa B, Díaz L, Gutiérrez C, Madrid N, et al. Correlation between different methods to measure microbial translocation and its association with immune activation in long‐term suppressed HIV‐1‐infected individuals. J Acquir Immune Defic Syndr. 1999;2013(64):149–53.
    1. Bengsch B, Seigel B, Flecken T, Wolanski J, Blum HE, Thimme R. Human Th17 cells express high levels of enzymatically active dipeptidylpeptidase IV (CD26). J Immunol. 2012;188:5438–47.
    1. Ohnuma K, Dang NH, Morimoto C. Revisiting an old acquaintance: CD26 and its molecular mechanisms in T cell function. Trends Immunol. 2008;29:295–301.
    1. Casrouge A, Decalf J, Ahloulay M, Lababidi C, Mansour H, Vallet‐Pichard A, et al. Evidence for an antagonist form of the chemokine CXCL10 in patients chronically infected with HCV. J Clin Invest. 2011;121:308–17.
    1. De Pasquale A, Ginaldi L, Limoncelli P, Quaglino D. Dipeptidyl amino peptidase IV cytochemistry in circulating lymphocytes from HIV‐I‐seropositive subjects. Acta Haematol. 1989;81:19–21.
    1. Blazquez MV, Madueño JA, Gonzalez R, Jurado R, Bachovchin WW, Peña J, et al. Selective decrease of CD26 expression in T cells from HIV‐1‐infected individuals. J Immunol. 1992;149:3073–7.
    1. Vanham G, Kestens L, De Meester I, Vingerhoets J, Penne G, Vanhoof G, et al. Decreased expression of the memory marker CD26 on both CD4+ and CD8+ T lymphocytes of HIV‐infected subjects. J Acquir Immune Defic Syndr. 1993;6:749–57.
    1. Gougeon ML, Lecoeur H, Callebaut C, Jacotot E, Dulioust A, Roué R, et al. Selective loss of the CD4+/CD26+ T‐cell subset during HIV infection. Res Immunol. 1996;147:5–8.
    1. Hosono O, Homma T, Kobayashi H, Munakata Y, Nojima Y, Iwamoto A, et al. Decreased dipeptidyl peptidase IV enzyme activity of plasma soluble CD26 and its inverse correlation with HIV‐1 RNA in HIV‐1 infected individuals. Clin Immunol. 1999;91:283–95.
    1. Ploquin MJ, Madec Y, Casrouge A, Huot N, Passaes C, Lécuroux C, et al. Elevated basal pre‐infection CXCL10 in plasma and in the small intestine after infection are associated with more rapid HIV/SIV disease onset. PLoS Pathog. 2016;12:e1005774.
    1. Euler Z, van Gils MJ, Boeser‐Nunnink BD, Schuitemaker H, van Manen D. Genome‐wide association study on the development of cross‐reactive neutralizing antibodies in HIV‐1 infected individuals. PLoS ONE. 2013;8:e54684.
    1. Boufassa F, Goujard C, Viard J‐P, Carlier R, Lefebvre B, Yeni P, et al. Immune deficiency could be an early risk factor for altered insulin sensitivity in antiretroviral‐naive HIV‐1‐infected patients: the ANRS COPANA cohort. Antivir Ther. 2012;17:91–100.
    1. Ghislain M, Bastard J‐P, Meyer L, Capeau J, Fellahi S, Gérard L, et al. Late antiretroviral therapy (ART) initiation is associated with long‐term persistence of systemic inflammation and metabolic abnormalities. PLoS ONE. 2015;10:e0144317.
    1. Liovat A‐S, Rey‐Cuillé M‐A, Lécuroux C, Jacquelin B, Girault I, Petitjean G, et al. Acute plasma biomarkers of T cell activation set‐point levels and of disease progression in HIV‐1 infection. PLoS ONE. 2012;7:e46143.
    1. Ghosn J, Pellegrin I, Goujard C, Deveau C, Viard J‐P, Galimand J, et al. HIV‐1 resistant strains acquired at the time of primary infection massively fuel the cellular reservoir and persist for lengthy periods of time. AIDS. 2006;20:159–70.
    1. Goujard C, Bonarek M, Meyer L, Bonnet F, Chaix M‐L, Deveau C, et al. CD4 cell count and HIV DNA level are independent predictors of disease progression after primary HIV type 1 infection in untreated patients. Clin Infect Dis. 2006;42:709–15.
    1. Laanani M, Ghosn J, Essat A, Melard A, Seng R, Gousset M, et al. Impact of the timing of initiation of antiretroviral therapy during primary HIV‐1 infection on the decay of cell‐associated HIV‐DNA. Clin Infect Dis. 2015;60:1715–21.
    1. Ngo‐Giang‐Huong N, Deveau C, Da Silva I, Pellegrin I, Venet A, Harzic M, et al. Proviral HIV‐1 DNA in subjects followed since primary HIV‐1 infection who suppress plasma viral load after one year of highly active antiretroviral therapy. AIDS. 2001;15:665–73.
    1. Diop OM, Gueye A, Dias‐Tavares M, Kornfeld C, Faye A, Ave P, et al. High levels of viral replication during primary simian immunodeficiency virus SIVagm infection are rapidly and strongly controlled in African green monkeys. J Virol. 2000;74:7538–47.
    1. Jacquelin B, Petitjean G, Kunkel D, Liovat A‐S, Jochems SP, Rogers KA, et al. Innate immune responses and rapid control of inflammation in African green monkeys treated or not with interferon‐alpha during primary SIVagm infection. PLoS Pathog. 2014;10:e1004241.
    1. Pandrea I, Kornfeld C, Ploquin MJ‐Y, Apetrei C, Faye A, Rouquet P, et al. Impact of viral factors on very early in vivo replication profiles in simian immunodeficiency virus SIVagm‐infected African green monkeys. J Virol. 2005;79:6249–59.
    1. Tejera‐Alhambra M, Casrouge A, de Andrés C, Ramos‐Medina R, Alonso B, Vega J, et al. Low DPP4 expression and activity in multiple sclerosis. Clin Immunol. 2014;150:170–83.
    1. Meissner EG, Decalf J, Casrouge A, Masur H, Kottilil S, Albert ML, et al. Dynamic changes of post‐translationally modified forms of CXCL10 and soluble DPP4 in HCV subjects receiving interferon‐free therapy. PLoS ONE. 2015;10:e0133236.
    1. Kornfeld C, Ploquin MJ‐Y, Pandrea I, Faye A, Onanga R, Apetrei C, et al. Antiinflammatory profiles during primary SIV infection in African green monkeys are associated with protection against AIDS. J Clin Invest. 2005;115:1082–91.
    1. Jacquelin B, Mayau V, Targat B, Liovat A‐S, Kunkel D, Petitjean G, et al. Nonpathogenic SIV infection of African green monkeys induces a strong but rapidly controlled type I IFN response. J Clin Invest. 2009;119:3544–55.
    1. Söderholm J, Waldenström J, Askarieh G, Pilli M, Bochud P‐Y, Negro F, et al. Impact of soluble CD26 on treatment outcome and hepatitis C virus‐specific T cells in chronic hepatitis C virus genotype 1 infection. PLoS ONE. 2013;8:e56991.
    1. Liang P‐I, Yeh B‐W, Li W‐M, Chan T‐C, Chang I‐W, Huang C‐N, et al. DPP4/CD26 overexpression in urothelial carcinoma confers an independent prognostic impact and correlates with intrinsic biological aggressiveness. Oncotarget. 2017;8:2995–3008.
    1. Kurktschiev D, Temelkova‐Kurktschiev T, Horn K, Schentke K. Successful immunomodulating in AIDS patients with ursodeoxycholic acid–a pilot study. Clin Exp Immunol. 1999;115:144–6.
    1. Garcia‐Tellez T, Huot N, Ploquin MJ, Rascle P, Jacquelin B, Müller‐Trutwin M. Non‐human primates in HIV research: achievements, limits and alternatives. Infect Genet Evol. 2016;46:324–32.
    1. Huot N, Jacquelin B, Garcia‐Tellez T, Rascle P, Ploquin MJ, Madec Y, et al. Natural killer cells migrate into and control simian immunodeficiency virus replication in lymph node follicles in African green monkeys. Nat Med. 2017;23:1277–86.
    1. Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14:667–85.
    1. Micci L, Ryan ES, Fromentin R, Bosinger SE, Harper JL, He T, et al. Interleukin‐21 combined with ART reduces inflammation and viral reservoir in SIV‐infected macaques. J Clin Invest. 2015;125:4497–513.
    1. Lee SA, Kim YR, Yang EJ, Kwon E‐J, Kim SH, Kang SH, et al. CD26/DPP4 levels in peripheral blood and T cells in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab. 2013;98:2553–61.
    1. Zheng T, Gao Y, Baskota A, Chen T, Ran X, Tian H. Increased plasma DPP4 activity is predictive of prediabetes and type 2 diabetes onset in Chinese over a four‐year period: result from the China National Diabetes and Metabolic Disorders Study. J Clin Endocrinol Metab. 2014;99:E2330–4.
    1. Grujic M, Matic IZ, Crnogorac MD, Velickovic AD, Kolundzija B, Cordero OJ, et al. Activity and expression of dipeptidyl peptidase IV on peripheral blood mononuclear cells in patients with early steroid and disease modifying antirheumatic drugs naïve rheumatoid arthritis. Clin Chem Lab Med. 2017;55:73–81.
    1. Stecca BA, Nardo B, Chieco P, Mazziotti A, Bolondi L, Cavallari A. Aberrant dipeptidyl peptidase IV (DPP IV/CD26) expression in human hepatocellular carcinoma. J Hepatol. 1997;27:337–45.
    1. Molica S, Digiesi G, Mirabelli R, Cutrona G, Antenucci A, Molica M, et al. Serum level of CD26 predicts time to first treatment in early B‐chronic lymphocytic leukemia. Eur J Haematol. 2009;83:208–14.
    1. Drucker DJ, Nauck MA. The incretin system: glucagon‐like peptide‐1 receptor agonists and dipeptidyl peptidase‐4 inhibitors in type 2 diabetes. Lancet. 2006;368:1696–705.
    1. Barreira da Silva R, Laird ME, Yatim N, Fiette L, Ingersoll MA, Albert ML. Dipeptidylpeptidase 4 inhibition enhances lymphocyte trafficking, improving both naturally occurring tumor immunity and immunotherapy. Nat Immunol 2015;16:850–8.
    1. Ragab D, Laird M, Duffy D, Casrouge A, Mamdouh R, Abass A, et al. CXCL10 antagonism and plasma sDPPIV correlate with increasing liver disease in chronic HCV genotype 4 infected patients. Cytokine. 2013;63:105–12.
    1. Sharma PK, Wong EB, Napier RJ, Bishai WR, Ndung'u T, Kasprowicz VO, et al. High expression of CD26 accurately identifies human bacteria‐reactive MR1‐restricted MAIT cells. Immunology 2015;145:443–53.
    1. Songok EM, Osero B, McKinnon L, Rono MK, Apidi W, Matey EJ, et al. CD26/dipeptidyl peptidase IV (CD26/DPPIV) is highly expressed in peripheral blood of HIV‐1 exposed uninfected female sex workers. Virol J. 2010;7:343.
    1. Songok EM, Luo M, Liang B, Mclaren P, Kaefer N, Apidi W, et al. Microarray analysis of HIV resistant female sex workers reveal a gene expression signature pattern reminiscent of a lowered immune activation state. PLoS ONE. 2012;7:e30048.
    1. Hartigan‐O'Connor DJ, Abel K, Van Rompay KKA, Kanwar B, McCune JM. SIV replication in the infected rhesus macaque is limited by the size of the preexisting TH17 cell compartment. Sci Transl Med. 2012;4:136ra69.
    1. Saez‐Cirion A, Jacquelin B, Barré‐Sinoussi F, Müller‐Trutwin M. Immune responses during spontaneous control of HIV and AIDS: what is the hope for a cure? Philos Trans R Soc Lond B Biol Sci. 2014;369:20130436.
    1. Mellors JW, Rinaldo CR, Gupta P, White RM, Todd JA, Kingsley LA. Prognosis in HIV‐1 infection predicted by the quantity of virus in plasma. Science. 1996;272:1167–70.

Source: PubMed

3
Abonnere