Skip to main content
Erschienen in: AIDS Research and Therapy 1/2018

Open Access 01.12.2018 | Review

Effects of HIV infection and ART on phenotype and function of circulating monocytes, natural killer, and innate lymphoid cells

verfasst von: Rose Nabatanzi, Stephen Cose, Moses Joloba, Sarah Rowland Jones, Damalie Nakanjako

Erschienen in: AIDS Research and Therapy | Ausgabe 1/2018

Abstract

HIV infection causes upregulation of markers of inflammation, immune activation and apoptosis of host adaptive, and innate immune cells particularly monocytes, natural killer (NK) and innate lymphoid cells (ILCs). Although antiretroviral therapy (ART) restores CD4 T-cell counts, the persistent aberrant activation of monocytes, NK and ILCs observed likely contributes to the incomplete recovery of T-cell effector functions. A better understanding of the effects of HIV infection and ART on the phenotype and function of circulating monocytes, NK, and ILCs is required to guide development of novel therapeutic interventions to optimize immune recovery.

Background

The human innate immune system is comprised of a complex network of cellular and soluble proteins that work together to provide the first-line of defense against common invading pathogens prior to involvement of the adaptive immune response [13]. Innate immune cells including monocytes, natural killer cells (NK), innate lymphoid cells (ILCs), and other antigen presenting cells (APCs) play a crucial role in the ushering in the adaptive arm of the immune response [4, 5]. In particular, monocytes are precursor cells to professional APCs involved in immune surveillance [6]. In addition, they have pattern-recognition receptors (PRRs) that detect conserved pathogen-associated molecular patterns (PAMPs) which lead to the induction of inflammatory responses that combat invading pathogens [7]. Natural killer cells produce cytokines; particularly interferon-gamma (IFN-ɣ) which activates phagocytic cells and primes APCs for interleukin 2 (IL-2) secretion thus shaping adaptive immunity towards a T helper 1 (Th1) response [8, 9]. ILCs rapidly secrete immunoregulatory cytokines which makes them provide protective immunity early on during infection [10] and also maintain intestinal homeostasis by directly regulating T cells through the presentation of peptide antigens on major histocompatibility complex II [11].
During HIV infection, the adaptive immune system is directly affected through the rapid infection of CD4 T-cells [12] but the effects on the innate immune system are more indirect through microbial translocation, inflammation, and immune activation [13]. Immune activation and inflammation cause a reduction in the numbers of monocytes, NK and ILCs, consequently leading to poor innate and adaptive immune responses, all which result in suboptimal response to infecting antigens [14].
Antiretroviral therapy (ART) suppresses HIV replication, restores CD4 T-cell numbers, reduces microbial translocation, inflammation, and aberrant T-cell activation [1517]. The net effect of this is the near restoration of the immune system to pre-infection status and control/prevention of opportunistic infections and other AIDS-associated ailments [18, 19]. Several studies have however demonstrated incomplete recovery of the adaptive immune responses including ours which showed lower CD4 T-cell proliferation among ART-treated adults (with CD4 counts restored to 500 cells/µl and more), relative to their HIV-negative counterparts [1921]. Innate immune cells; in particular NK cells, ILCs and monocytes, participate in the initiation and development of adaptive immune responses although little is known about their recovery during ART. This review discusses the recovery of monocytes, NK and ILCs during ART, because of their respective contributions to the regulation of the adaptive immune response.
Figure 1 summarises the effects of HIV infection and ART on monocytes, NK cells, ILCs, and other innate cells. Persistent inflammation and activation of monocytes, NK cells, and ILCs is likely associated with the persistent T-cell activation and impaired effector functions observed among ART-treated adults [2022]. There is likely a unique phenomenon of innate immune cell recovery during ART, among residents in sub-Saharan Africa (SSA) where several endemic infections activate the immune systems. A better understanding of innate cell dysfunctions and their effects on the adaptive responses during ART would guide the development of innovative therapeutic intervention to optimize recovery of host immune responses.

HIV infection and innate immune cells

Monocytes

In the first few weeks of HIV infection, there is a massive accumulation of CD8 T-cells and a massive depletion of CD4 T-cells in the gut, followed by increased gut permeability and translocation of microbial products into circulation [23, 24]. Microbial translocation contributes to increased monocyte activation as evidenced by the rapid shift in the circulating monocyte pool from the classical phagocytic monocytes (CD14++CD16) to the intermediate inflammatory monocyte subpopulation (CD14++CD16+) in the first 2 weeks of HIV infection [25]. Subsequently, monocyte subsets are disrupted, leading to suboptimal effector functions of phagocytosis, intracellular killing, chemotaxis and cytokine production [26]. HIV infection, both through direct infection and indirectly through microbial translocation, leads to monocyte activation and aberrant release of pro-inflammatory cytokines including TNF-α, IL-1β and IL-6, thereby activating the immune system [17]. In addition to aberrant cytokine production, HIV-associated monocyte activation leads to increased release of chemokines, leading to non-specific movement of monocytes into various tissue sites [23]. Direct HIV infection of the monocytes down regulates MHCII expression, inhibits MHCII-antigen complex formation and reduces the monocyte ability to take up antigens for processing and presentation to T cells [27]. In a study conducted in Beijing, Chen et al. [28] demonstrated that acute HIV-1 infected individuals had significantly increased proportions of inflammatory monocyte subsets and upregulated expression of the HLA-DR and CD163 receptors when compared with HIV negative individuals. These acquired defects in monocyte function cause the inability of monocytes to present antigens [27].
We postulate that the observed increase in inflammatory monocytes and immune activation markers could further impair monocyte responsiveness to antigens making the HIV infected individuals more susceptible to opportunistic infections. After several months to years of HIV infection, viral load levels gradually increase, while CD4 T-cells continue to reduce in number and function [2931]. Similarly, there is dysregulation of monocyte subsets with higher populations of inflammatory (CD14+CD16+) monocytes than phagocytic (CD14+CD16) populations. Monocytes in circulation become functionally anergic due to continued activation and high inflammatory status [32]. Untreated, individuals with chronic HIV-1 infection continue to have increased proportions of both the intermediate and non-classical monocytes subsets [33]. Moreover, levels of expression of CD163, a marker of activation on inflammatory monocytes remains significantly higher among individuals with chronic HIV-1-infection than HIV negative controls [34]. Protracted expression of the activated phenotype of monocyte subsets has a direct association with disease progression [33].
ART down regulates the excessive production of cytokines by the inflammatory monocytes thereby reducing the levels of immune activation and inflammation [17]. In a review by Burdo et al. [35], it was observed that ART initiation within the first year of HIV infection reduced monocyte activation, as evidenced by a reduction in expression of activation marker CD163 and absolute numbers of inflammatory monocytes. Similarly, markers of microbial translocation [lipopolysaccharide protein (LPS), IL-6, 16S ribosomal DNA and soluble CD14], and inflammatory markers such as d-dimer and interferon-α declined with ART initiation [33, 36]. Although a lot of immune functions appear to be recovered during ART, some monocyte dysfunctions persist. After 1 year of therapy, ART-treated adults were reported to still have elevated levels of the inflammatory monocyte subset (CD14++CD16+) and a downregulated expression of phagocytic monocyte subset (CD14++CD16), resulting in the reduced ability of monocytes to process and present antigens to T cells [34, 37]. Similarly, phagocytic activity and oxidative burst of neutrophils and monocytes remained impaired among HIV-1 infected patients, in Athens general hospital after 3 months of ART [26, 38]. However, there is paucity of data on monocyte activation and functional recovery beyond 2 years of ART, particularly in sub-Saharan Africa (SSA) where monocyte frequency and functional recovery has not been widely studied in HIV treatment cohorts. Given the increasing numbers of individuals receiving ART, for 7 years and over, a better understanding of the effects of long-term ART on inflammation and monocyte activation would be relevant to inform innovations against chronic inflammation and its complications among adults living with HIV.

Natural killer cells

Natural killer (NK) cells have an important role in controlling acute HIV infection, through rapid division and production of huge amounts of IFN-γ cytokine [8]. Strong NK cell activity and cytotoxic receptor expression are associated with preservation of CD4 T cells and lower viral set point [39]. HIV infection is associated with several changes in the NK cell compartment, including phenotypic and functional abnormalities that contribute to difficulty in the control of HIV progression [40]. Evidence of dysfunctional NK cell populations has been revealed by studies in the nonhuman primate model which have demonstrated anergic NK cell accumulation in lymph nodes in SIV infection [41].
In humans, acute HIV infection generally causes activation and expansion of the whole pool of NK cells [42], with abnormal distribution of the NK cell subsets. Pro-inflammatory NKCD56bright populations are reduced, while the cytolytic CD56dimCD16pos NK cell and dysfunctional CD56negCD16pos NK cells are increased in HIV positive people compared to HIV negative individuals [43, 44]. HIV infection reduces expression of the natural cytotoxicity receptors (NCR), NKp30, NKp44 and NKp46 [39, 45], which are essential in the containment and clearance of HIV virus. Evidence has further suggested that acute HIV infection activates the upregulation of stress ligands for cytotoxicity receptors including NKG2D which leads to lysis and cell death [46].
With continued viral replication, the CD56dimCD16pos NK cell subset previously expanded in acute infection drop in numbers and function, as demonstrated by reduced CD107a expression and cytokine secretion [47]. The reduced numbers of cytolytic and cytokine producing NK cells would suggest that HIV infected patients with chronic disease remain susceptible to many infections especially those of viral origin [42, 48]. Evidence of reduced cytokine producing NK cells was further demonstrated in a rural Ugandan cohort, where chronically infected HIV individuals had lower expression of the NKG2A+CD57+CD56dim subset in HIV infected group than the HIV negative controls [49]. In addition to down-regulated cytokine production, HIV infection causes a reduced ability of NK cells to perform ADCC due to a reduction in the number of the cytolytic CD56dimCD16+NK cells population [50] and a reduction in the intracellular stores of perforin and granzyme A [51].
Several discrepant results have been reported on the recovery of NK cells with ART.
ART has been shown to restore NK cell numbers with a mature phenotype in HIV-infected individuals, although defects of subset distribution and impaired ability to produce IFN-ɣ cytokine persist [44, 52, 53]. Frias et al. [54] reported incomplete recovery of NK cell subsets after 3 years of ART in Spain, in spite of undetectable viral load and an outstanding increase in the CD4 count to levels above 500 cell/µl. On the contrary, Mavilio et al. and Luo et al. [40, 55] reported complete recovery of NK cell subsets and functional profiles after 2 years of ART when compared with HIV negative individuals. In Ottawa at the immunodeficiency clinic, NK cell cytolytic activity was greatly reduced to levels similar to those among HIV negative people, after 1 year of ART [56]. In a Ugandan cohort, we demonstrated increased pro-inflammatory CD56bright NK cells that were associated with suboptimal immune recovery despite 4 years of suppressive therapy [57]. Given that most of the evidence on the recovery of NK cells is from individuals after a short duration of ART, there is need to explore recovery of NK cell function after longer durations of ART. This is particularly important for sub-Saharan Africa which has more than 10 million people receiving ART majority of whom have been on treatment for more than 10 years [58].

Innate lymphoid cells (ILCs)

Innate lymphoid cells are a group of innate immune cells that belong to the lymphoid lineage but do not respond in an antigen-specific manner, because of their lack of a B or T cell receptor [59]. These cells are subdivided into ILC1, ILC2 and ILC3 and these mirror the CD4 T helper cells TH1, TH2, and TH17 cells in the cytokines they produce. ILCs are mainly found at mucosal surfaces where they act as gatekeepers to invading infectious agents, including HIV [60]. ILCs rapidly secrete immunoregulatory cytokines which makes them provide protective immunity early on during infection [61]. Studies done in non-human primates revealed that ILC populations in the gut mucosa are significantly reduced in numbers due to an increase in cytotoxicity and inflammatory cytokine production by both ILCs and NK cells during acute Simian immunodeficiency viruses (SIV) infection. The reduction in ILC numbers contributes to the massive apoptosis and dysregulation in the gut-associated lymphoid tissue (GALT) [62, 63]. Xu et al. [64] showed that IL-17 producing ILC populations were drastically reduced in acute SIV infection, especially in the jejunum.
In humans, acute HIV infection has been associated with lymphoid tissue destruction of gut mucosa and further causes upregulation of genes associated with ILCs cell death, as evidenced by the depletion of ILCs both in blood and gut tissues of HIV-1 infected patients [63]. The destruction of lymphoid tissue cells has been associated with microbial translocation, immune activation and disease progression in both ART-treated and untreated individuals [65]. In a study by Kløverpris et al. among individuals with acute HIV infection in South Africa, it was demonstrated that all three subsets of ILCs were massively depleted from peripheral blood 7–14 days after HIV infection and these did not increase with viral load decreasing [65, 66].
In chronic HIV infection, ILC3s are further depleted and the depletion was attributed to the presence of excessive production of type 1 interferons by the plasmacytoid dendritic cells [67]. Although ILCs are depleted irreversibly from peripheral blood and the mucosal tissues, Mudd et al. [67] demonstrated that ILCs in tonsillar tissue are not significantly altered, meaning that ILC depletion is not generalised but rather compartmentalised and with continued therapy they may be redistributed back in peripheral blood and mucosal tissues.
Initiation of ART during acute HIV infection, preserves ILC numbers if it is initiated before peak viremia [66]. However, ART initiation during chronic HIV disease seems to have little effect on recovery of ILC numbers; with circulating ILC1s and ILC2s remaining significantly depleted and incomplete reconstitution of circulating ILC3s even with 2 years of ART [65, 66]. Kramer et al. studied ILC distribution in the gut and observed that despite effective use of ART, ILCs in HIV infected individuals remain dysregulated compared to their HIV negative counterparts. This lack of recovery of ILC distribution may contribute to the loss of intestinal barrier integrity and immune activation [66, 68, 69]. It is likely that individuals with persistent ILC dysfunction remain with limited mucosal protection and subsequently high risk of bacterial infections, autoimmune diseases and allergic infections due to the subsequent limitations in TH1, TH2 and TH17 functions that are mirrored by ILC1, ILC2 and ILC3 phenotypes, respectively [68].

Consequences and clinical implications of persistent dysfunction of innate immune cells during ART

Innate immune cells and IRIS

ART generally leads to viral suppression, improvement of immune function, and better outcomes for many HIV positive individuals. Up to 10–25% of ART-treated individuals [70, 71] may develop the immune reconstitution inflammatory syndrome (IRIS) during the first months to years of HIV treatment. IRIS is a paradoxical inflammatory syndrome resulting from increased host immune responses to pre-existing opportunistic pathogens, including Mycobacterium avium complex, M. tuberculosis, Cryptococcus neoformans, Cytomegalovirus, JC virus, Pneumocystis jirovecii, Herpes zoster (VZV), and hepatitis B, as a result of CD4 T-cell restoration and interferon gamma production during the first months of ART [72]. The contribution of the different innate immune cells to IRIS has been documented by different groups; for example, in a Ugandan cohort, Tran et al. [73, 74] reported that monocyte-associated biological processes and functions were disturbed in TB-IRIS patients (after 2 weeks of ART), with dysregulation in both anti- and pro-inflammatory processes in monocytes. Andrade et al. [75] evaluated soluble biomarkers of inflammation and monocyte activation in patients who had been on ART for 12 weeks with TB-HIV co-infection from India and South Africa and found increased plasma levels of sCD14 and sCD163 pre vs post IRIS which are strong indicators of monocyte activation and predictors of death in TB-IRIS patients. Natural killer cells also exhibit phenotypic and functional differences in patients who develop IRIS relative to those who do not. At a Themba Lethu clinic Johannesburg, patients who developed IRIS had significantly higher levels of NK-cell degranulation before ART initiation [76] and exhibited high immune activation levels as demonstrated by the elevated levels of CD69 and HLA-DR [77]. Increased NK cell degranulation can cause lysis of cells infected with antigens, thereby increasing the circulating antigen load in these patients and contributing to the observed IRIS [76]. Given the propagating role of monocyte activation in IRIS processes, it is likely that therapeutic interventions to minimise monocyte activation might indirectly modify the risk and severity of IRIS among ART-treated adults.

Persistent immune activation and non-AIDS complications

Aberrant activation of the innate immune system is persistent despite ART [78], and it could be directly due to replicating HIV virus or indirectly through co-infections including subclinical Cytomegalovirus (CMV) infection [79]. Innate immune activation can be due to HIV directly infecting the monocytes/macrophages and dendritic cells or indirectly through HIV gene products like envelop proteins of gp120 and Nef that cause activation of lymphocytes and macrophages to produce pro-inflammatory cytokines and chemokines [80]. Evidence of indirect immune activation suggests that persistent leakage of lipopolysaccharide (LPS) into blood circulation [36] causes monocyte activation [81].
Persistent activation of innate immune cells is associated with the heightened production of pro-inflammatory cytokines (IL-1β, TNFα and IL-6) which cause T-cell activation. T-cell activation subsequently increases intracellular NF-κB levels which enhances the transcription of integrated virus and production of new virions that further infect more cells [82]. Activation of T-cells promotes T-cell depletion through upregulation of apoptosis, ADCC, and by-stander killing; all of which are functions of innate immune cells [3]. Reduced numbers of innate monocytes, NK and ILCs, consequently lead to poor innate and adaptive immune responses causing suboptimal response to infecting antigens [14]. Moreover, persistent inflammation and activation have been associated with fatal non-AIDS illnesses such as cardiovascular diseases, malignancies and organ damage among adults aging with HIV [8385]. The high levels of inflammation and immune activation associated with chronic HIV disease, despite ART, contribute to accelerated immune aging and increase the risk of non-AIDS illnesses including cardiovascular diseases [86, 87], cataracts [88], malignancies [89, 90], bone demineralization [91], renal disease [92] and cognitive decline [93]) among HIV-positive adults relative to their HIV-negative counterparts. We, therefore, postulate that strategies to downgrade innate immune cell activation and associated dysfunctions could modify the magnitude, duration, and systemic complications of the aberrant immune activation associated with HIV chronic disease.

Conclusions

HIV infection disrupts phenotypes and functions of monocytes, NK cells and ILCs, and subsequently the related adaptive host immune responses. ART restores some phenotypic and functional abnormalities associated with HIV infection, although persistent disruption of phenotypes and function of monocytes, NK cells, and innate lymphoid cells have been observed among populations of ART-treated adults. A further understanding of specific persistent innate immune cell phenotypic and functional abnormalities during ART is required to inform innovations in immune modulation interventions to optimize recovery of both innate and adaptive immune system. Similarly, a further understanding of the drivers of persistent immune activation is required to inform strategic therapeutic interventions to minimize its complications, particularly in sub-Saharan Africa where other infectious causes of immune activation such as malaria, tuberculosis and helminthic infections are still endemic.

Authors’ contributions

RN, SC, MJ, SRJ, DN contributed to the conceptualization and execution of the review. RN and DN drafted the manuscript. All authors reviewed manuscript for publication. All authors read and approved the final manuscript.

Acknowledgements

The authors also acknowledge funding from the Alliance for Global Health and Science at the University of California, Berkeley, USA and the Wheeler Center for Emerging and Neglected Diseases, University of California, Berkeley, USA that funded our research to further understand Natural Killer cells and Innate immune cells during ART.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.
Not applicable.
Not applicable.

Funding

This work was supported through the DELTAS Africa Initiative [Grant# 107743/Z/15/Z], that funded Damalie Nakanjako and Rose Nabatanzi through a group leader award. The DELTAS Africa Initiative is an independent funding scheme of the African Academy of Sciences (AAS)’s Alliance for Accelerating Excellence in Science in Africa (AESA) and supported by the New Partnership for Africa’s Development Planning and Coordinating Agency (NEPAD Agency) with funding from the Wellcome Trust [Grant #107743/Z/15/Z] and the UK government. The views expressed in this publication are those of the author(s) and not necessarily those of AAS, NEPAD Agency, Wellcome Trust or the UK government.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
3.
Zurück zum Zitat Ferreira C, et al. Differential survival of naive CD4 and CD8 T cells. J Immunol. 2000;165(7):3689–94.CrossRefPubMed Ferreira C, et al. Differential survival of naive CD4 and CD8 T cells. J Immunol. 2000;165(7):3689–94.CrossRefPubMed
6.
Zurück zum Zitat Yona S, Jung S. Monocytes: subsets, origins, fates and functions. Curr Opin Hematol. 2010;17(1):53–9.CrossRefPubMed Yona S, Jung S. Monocytes: subsets, origins, fates and functions. Curr Opin Hematol. 2010;17(1):53–9.CrossRefPubMed
7.
Zurück zum Zitat Ziegler-Heitbrock L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007;81(3):584–92.CrossRefPubMed Ziegler-Heitbrock L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007;81(3):584–92.CrossRefPubMed
8.
Zurück zum Zitat Terunuma H, et al. Potential role of NK cells in the induction of immune responses: implications for NK cell–based immunotherapy for cancers and viral infections. Int Rev Immunol. 2008;27(3):93–110.CrossRefPubMed Terunuma H, et al. Potential role of NK cells in the induction of immune responses: implications for NK cell–based immunotherapy for cancers and viral infections. Int Rev Immunol. 2008;27(3):93–110.CrossRefPubMed
9.
Zurück zum Zitat Vivier E, Ugolini S. NK cells: receptors and functions. Nat Rev Immunol. 2010;2:18. Vivier E, Ugolini S. NK cells: receptors and functions. Nat Rev Immunol. 2010;2:18.
10.
Zurück zum Zitat Leavy O. Innate-like lymphocytes: will the real ILC1 please stand up? Nat Rev Immunol. 2013;13(2):67.CrossRefPubMed Leavy O. Innate-like lymphocytes: will the real ILC1 please stand up? Nat Rev Immunol. 2013;13(2):67.CrossRefPubMed
11.
12.
Zurück zum Zitat Douek DC, et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature. 2002;417(6884):95.CrossRefPubMed Douek DC, et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature. 2002;417(6884):95.CrossRefPubMed
13.
Zurück zum Zitat Appay V, Kelleher AD. Immune activation and immune aging in HIV infection. Curr Opin HIV AIDS. 2016;11(2):242–9.CrossRefPubMed Appay V, Kelleher AD. Immune activation and immune aging in HIV infection. Curr Opin HIV AIDS. 2016;11(2):242–9.CrossRefPubMed
16.
Zurück zum Zitat Schuetz A, et al. Early ART initiation prevents disruption of the mucosal barrier and subsequent T-cell activation. CROI Abstract. 2014. Schuetz A, et al. Early ART initiation prevents disruption of the mucosal barrier and subsequent T-cell activation. CROI Abstract. 2014.
17.
Zurück zum Zitat Amirayan-Chevillard N, et al. Impact of highly active anti-retroviral therapy (HAART) on cytokine production and monocyte subsets in HIV-infected patients. Clin Exp Immunol. 2000;120(1):107–12.CrossRefPubMedPubMedCentral Amirayan-Chevillard N, et al. Impact of highly active anti-retroviral therapy (HAART) on cytokine production and monocyte subsets in HIV-infected patients. Clin Exp Immunol. 2000;120(1):107–12.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Funderburg NT, et al. Dynamics of immune reconstitution and activation markers in HIV+ treatment-naive patients treated with raltegravir, tenofovir disoproxil fumarate and emtricitabine. PLoS ONE. 2013;8(12):e83514.CrossRefPubMedPubMedCentral Funderburg NT, et al. Dynamics of immune reconstitution and activation markers in HIV+ treatment-naive patients treated with raltegravir, tenofovir disoproxil fumarate and emtricitabine. PLoS ONE. 2013;8(12):e83514.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Lederman MM. Immune restoration and CD4+ T-cell function with antiretroviral therapies. Aids. 2001;15:S11–5.CrossRefPubMed Lederman MM. Immune restoration and CD4+ T-cell function with antiretroviral therapies. Aids. 2001;15:S11–5.CrossRefPubMed
20.
Zurück zum Zitat Nabatanzi R, et al. Low antigen-specific CD4 T-cell immune responses despite normal absolute CD4 counts after long-term antiretroviral therapy an African cohort. Immunol Lett. 2014;162(2):264–72.CrossRefPubMedPubMedCentral Nabatanzi R, et al. Low antigen-specific CD4 T-cell immune responses despite normal absolute CD4 counts after long-term antiretroviral therapy an African cohort. Immunol Lett. 2014;162(2):264–72.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Nakanjako D, et al. Impaired T-cell proliferation among HAART-treated adults with suboptimal CD4 recovery in an African cohort. BMC Immunol. 2013;14(1):26.CrossRefPubMedPubMedCentral Nakanjako D, et al. Impaired T-cell proliferation among HAART-treated adults with suboptimal CD4 recovery in an African cohort. BMC Immunol. 2013;14(1):26.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Nakanjako D, et al. High T-cell immune activation and immune exhaustion among individuals with suboptimal CD4 recovery after 4 years of antiretroviral therapy in an African cohort. BMC Infect Dis. 2011;11(1):43.CrossRefPubMedPubMedCentral Nakanjako D, et al. High T-cell immune activation and immune exhaustion among individuals with suboptimal CD4 recovery after 4 years of antiretroviral therapy in an African cohort. BMC Infect Dis. 2011;11(1):43.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Ancuta P, et al. Microbial translocation is associated with increased monocyte activation and dementia in AIDS patients. PLoS ONE. 2008;3(6):e2516.CrossRefPubMedPubMedCentral Ancuta P, et al. Microbial translocation is associated with increased monocyte activation and dementia in AIDS patients. PLoS ONE. 2008;3(6):e2516.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013;21(1):6–13.CrossRefPubMed Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013;21(1):6–13.CrossRefPubMed
25.
Zurück zum Zitat Kim WK, et al. Monocyte heterogeneity underlying phenotypic changes in monocytes according to SIV disease stage. J Leukoc Biol. 2010;87(4):557–67.CrossRefPubMed Kim WK, et al. Monocyte heterogeneity underlying phenotypic changes in monocytes according to SIV disease stage. J Leukoc Biol. 2010;87(4):557–67.CrossRefPubMed
26.
Zurück zum Zitat Sassé T, et al. Monocytes and their role in human immunodeficiency virus pathogenesis. Am J Infect Dis. 2012;8(2):92.CrossRef Sassé T, et al. Monocytes and their role in human immunodeficiency virus pathogenesis. Am J Infect Dis. 2012;8(2):92.CrossRef
28.
Zurück zum Zitat Chen P, et al. Perturbations of monocyte subsets and their association with T helper cell differentiation in acute and chronic HIV-1-infected patients. Front Immunol. 2017;8:272.PubMedPubMedCentral Chen P, et al. Perturbations of monocyte subsets and their association with T helper cell differentiation in acute and chronic HIV-1-infected patients. Front Immunol. 2017;8:272.PubMedPubMedCentral
29.
Zurück zum Zitat Jansen CA, et al. Analysis of the effect of highly active antiretroviral therapy during acute HIV-1 infection on HIV-specific CD4 T cell functions. Aids. 2005;19(11):1145–54.CrossRefPubMed Jansen CA, et al. Analysis of the effect of highly active antiretroviral therapy during acute HIV-1 infection on HIV-specific CD4 T cell functions. Aids. 2005;19(11):1145–54.CrossRefPubMed
32.
Zurück zum Zitat Poovan K, Ipp H, Glashoff RH. Monocytes in chronic HIV-1 infection: putative gut homing markers and their relationship with immune activation. AIDS Res Hum Retroviruses. 2014;30(S1):A128.CrossRef Poovan K, Ipp H, Glashoff RH. Monocytes in chronic HIV-1 infection: putative gut homing markers and their relationship with immune activation. AIDS Res Hum Retroviruses. 2014;30(S1):A128.CrossRef
33.
34.
Zurück zum Zitat Han J, et al. CD14highCD16+ rather than CD14lowCD16+ monocytes correlate with disease progression in chronic HIV-infected patients. JAIDS J Acquir Immune Defic Syndr. 2009;52(5):553–9.CrossRefPubMed Han J, et al. CD14highCD16+ rather than CD14lowCD16+ monocytes correlate with disease progression in chronic HIV-infected patients. JAIDS J Acquir Immune Defic Syndr. 2009;52(5):553–9.CrossRefPubMed
36.
Zurück zum Zitat Marchetti G, Tincati C, Silvestri G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin Microbiol Rev. 2013;26(1):2–18.CrossRefPubMedPubMedCentral Marchetti G, Tincati C, Silvestri G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin Microbiol Rev. 2013;26(1):2–18.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007;82(2):244–52.CrossRefPubMed Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007;82(2):244–52.CrossRefPubMed
38.
Zurück zum Zitat Michailidis C, et al. Impaired phagocytosis among patients infected by the human immunodeficiency virus: implication for a role of highly active anti-retroviral therapy. Clin Exp Immunol. 2012;167(3):499–504.CrossRefPubMedPubMedCentral Michailidis C, et al. Impaired phagocytosis among patients infected by the human immunodeficiency virus: implication for a role of highly active anti-retroviral therapy. Clin Exp Immunol. 2012;167(3):499–504.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat De Maria A, et al. The impaired NK cell cytolytic function in viremic HIV-1 infection is associated with a reduced surface expression of natural cytotoxicity receptors (NKp46, NKp30 and NKp44). Eur J Immunol. 2003;33(9):2410–8.CrossRefPubMed De Maria A, et al. The impaired NK cell cytolytic function in viremic HIV-1 infection is associated with a reduced surface expression of natural cytotoxicity receptors (NKp46, NKp30 and NKp44). Eur J Immunol. 2003;33(9):2410–8.CrossRefPubMed
40.
Zurück zum Zitat Mavilio D, et al. Natural killer cells in HIV-1 infection: dichotomous effects of viremia on inhibitory and activating receptors and their functional correlates. Proc Natl Acad Sci. 2003;100(25):15011–6.CrossRefPubMedPubMedCentral Mavilio D, et al. Natural killer cells in HIV-1 infection: dichotomous effects of viremia on inhibitory and activating receptors and their functional correlates. Proc Natl Acad Sci. 2003;100(25):15011–6.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Schafer JL, et al. Accumulation of cytotoxic CD16+ NK cells in simian immunodeficiency virus-infected lymph nodes associated with in situ differentiation and functional anergy. J Virol. 2015;89(13):6887–94.CrossRefPubMedPubMedCentral Schafer JL, et al. Accumulation of cytotoxic CD16+ NK cells in simian immunodeficiency virus-infected lymph nodes associated with in situ differentiation and functional anergy. J Virol. 2015;89(13):6887–94.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Iannello A, et al. Antiviral NK cell responses in HIV infection: I. NK cell receptor genes as determinants of HIV resistance and progression to AIDS. J Leukoc Biol. 2008;84(1):1–26.CrossRefPubMed Iannello A, et al. Antiviral NK cell responses in HIV infection: I. NK cell receptor genes as determinants of HIV resistance and progression to AIDS. J Leukoc Biol. 2008;84(1):1–26.CrossRefPubMed
43.
Zurück zum Zitat Alter G, et al. Sequential deregulation of NK cell subset distribution and function starting in acute HIV-1 infection. Blood. 2005;106(10):3366–9.CrossRefPubMed Alter G, et al. Sequential deregulation of NK cell subset distribution and function starting in acute HIV-1 infection. Blood. 2005;106(10):3366–9.CrossRefPubMed
44.
46.
Zurück zum Zitat Bayigga L, et al. High CD56++ CD16-natural killer (NK) cells among suboptimal immune responders after four years of suppressive antiretroviral therapy in an African adult HIV treatment cohort. BMC Immunol. 2014;15(1):1.CrossRef Bayigga L, et al. High CD56++ CD16-natural killer (NK) cells among suboptimal immune responders after four years of suppressive antiretroviral therapy in an African adult HIV treatment cohort. BMC Immunol. 2014;15(1):1.CrossRef
47.
Zurück zum Zitat De Maria A, Moretta L. NK cell function in HIV-1 infection. Curr HIV Res. 2008;6(5):433–40.CrossRefPubMed De Maria A, Moretta L. NK cell function in HIV-1 infection. Curr HIV Res. 2008;6(5):433–40.CrossRefPubMed
48.
Zurück zum Zitat Jensen SS, et al. Restoration of the NK cells ability to mediate ADCC in HIV-1 positives after six months of HAART can be explained by normalization of their phenotype. AIDS Res Hum Retroviruses. 2014;30(S1):A126.CrossRef Jensen SS, et al. Restoration of the NK cells ability to mediate ADCC in HIV-1 positives after six months of HAART can be explained by normalization of their phenotype. AIDS Res Hum Retroviruses. 2014;30(S1):A126.CrossRef
49.
Zurück zum Zitat Naluyima P, et al. Impaired natural killer cell responses are associated with loss of the highly activated NKG2A+ CD57+ CD56dim subset in HIV-1 subtype D infection in Uganda. AIDS (London, England). 2014;28(9):1273.CrossRef Naluyima P, et al. Impaired natural killer cell responses are associated with loss of the highly activated NKG2A+ CD57+ CD56dim subset in HIV-1 subtype D infection in Uganda. AIDS (London, England). 2014;28(9):1273.CrossRef
50.
Zurück zum Zitat Sips M, et al. Altered distribution of mucosal NK cells during HIV infection. Mucosal Immunol. 2012;5(1):30–40.CrossRefPubMed Sips M, et al. Altered distribution of mucosal NK cells during HIV infection. Mucosal Immunol. 2012;5(1):30–40.CrossRefPubMed
52.
Zurück zum Zitat Azzoni L, et al. Sustained impairment of IFN-γ secretion in suppressed HIV-infected patients despite mature NK cell recovery: evidence for a defective reconstitution of innate immunity. J Immunol. 2002;168(11):5764–70.CrossRefPubMed Azzoni L, et al. Sustained impairment of IFN-γ secretion in suppressed HIV-infected patients despite mature NK cell recovery: evidence for a defective reconstitution of innate immunity. J Immunol. 2002;168(11):5764–70.CrossRefPubMed
53.
Zurück zum Zitat Chehimi J, et al. Baseline viral load and immune activation determine the extent of reconstitution of innate immune effectors in HIV-1-infected subjects undergoing antiretroviral treatment. J Immunol. 2007;179(4):2642–50.CrossRefPubMed Chehimi J, et al. Baseline viral load and immune activation determine the extent of reconstitution of innate immune effectors in HIV-1-infected subjects undergoing antiretroviral treatment. J Immunol. 2007;179(4):2642–50.CrossRefPubMed
54.
Zurück zum Zitat Frias M, et al. Persistence of pathological distribution of NK cells in HIV-infected patients with prolonged use of HAART and a sustained immune response. PLoS ONE. 2015;10(3):e0121019.CrossRefPubMedPubMedCentral Frias M, et al. Persistence of pathological distribution of NK cells in HIV-infected patients with prolonged use of HAART and a sustained immune response. PLoS ONE. 2015;10(3):e0121019.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Luo Z, et al. Increased natural killer cell activation in HIV-infected immunologic non-responders correlates with CD4+ T cell recovery after antiretroviral therapy and viral suppression. PLoS ONE. 2017;12(1):e0167640.CrossRefPubMedPubMedCentral Luo Z, et al. Increased natural killer cell activation in HIV-infected immunologic non-responders correlates with CD4+ T cell recovery after antiretroviral therapy and viral suppression. PLoS ONE. 2017;12(1):e0167640.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Parato KG, et al. Normalization of natural killer cell function and phenotype with effective anti-HIV therapy and the role of IL-10. Aids. 2002;16(9):1251–6.CrossRefPubMed Parato KG, et al. Normalization of natural killer cell function and phenotype with effective anti-HIV therapy and the role of IL-10. Aids. 2002;16(9):1251–6.CrossRefPubMed
57.
Zurück zum Zitat Bayigga L, et al. High CD56++ CD16-natural killer (NK) cells among suboptimal immune responders after four years of suppressive antiretroviral therapy in an African adult HIV treatment cohort. BMC Immunol. 2014;15(1):2.CrossRefPubMedPubMedCentral Bayigga L, et al. High CD56++ CD16-natural killer (NK) cells among suboptimal immune responders after four years of suppressive antiretroviral therapy in an African adult HIV treatment cohort. BMC Immunol. 2014;15(1):2.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Flynn AG, et al. Socioeconomic position and ten-year survival and virologic outcomes in a Ugandan HIV cohort receiving antiretroviral therapy. PLoS ONE. 2017;12(12):e0189055.CrossRefPubMedPubMedCentral Flynn AG, et al. Socioeconomic position and ten-year survival and virologic outcomes in a Ugandan HIV cohort receiving antiretroviral therapy. PLoS ONE. 2017;12(12):e0189055.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells—how did we miss them? Nat Rev Immunol. 2013;13(2):75–87.CrossRefPubMed Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells—how did we miss them? Nat Rev Immunol. 2013;13(2):75–87.CrossRefPubMed
60.
Zurück zum Zitat Almeida F, Belz G. Innate lymphoid cells: models of plasticity for immune homeostasis and rapid responsiveness in protection. Mucosal Immunol. 2016;9(5):1103–12.CrossRefPubMed Almeida F, Belz G. Innate lymphoid cells: models of plasticity for immune homeostasis and rapid responsiveness in protection. Mucosal Immunol. 2016;9(5):1103–12.CrossRefPubMed
61.
Zurück zum Zitat Brenchley JM, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–71.CrossRefPubMed Brenchley JM, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–71.CrossRefPubMed
62.
63.
Zurück zum Zitat Reeves RK, et al. Depletion of lamina propria innate lymphoid cells in simian immunodeficiency virus infection. AIDS Res Hum Retroviruses. 2014;30(12):1160–1.CrossRefPubMed Reeves RK, et al. Depletion of lamina propria innate lymphoid cells in simian immunodeficiency virus infection. AIDS Res Hum Retroviruses. 2014;30(12):1160–1.CrossRefPubMed
64.
Zurück zum Zitat Xu H, et al. IL-17-producing innate lymphoid cells are restricted to mucosal tissues and are depleted in SIV-infected macaques. Mucosal Immunol. 2012;5(6):658–69.CrossRefPubMedPubMedCentral Xu H, et al. IL-17-producing innate lymphoid cells are restricted to mucosal tissues and are depleted in SIV-infected macaques. Mucosal Immunol. 2012;5(6):658–69.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Kløverpris HN, et al. Innate lymphoid cells are depleted in HIV infection. AIDS Res Hum Retroviruses. 2014;30(S1):A14.CrossRef Kløverpris HN, et al. Innate lymphoid cells are depleted in HIV infection. AIDS Res Hum Retroviruses. 2014;30(S1):A14.CrossRef
66.
Zurück zum Zitat Kløverpris HN, et al. Innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression. Immunity. 2016;44(2):391–405.CrossRefPubMed Kløverpris HN, et al. Innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression. Immunity. 2016;44(2):391–405.CrossRefPubMed
67.
Zurück zum Zitat Mudd JC, Brenchley JM. ILC you later: early and irreparable loss of innate lymphocytes in HIV infection. Immunity. 2016;44(2):216–8.CrossRefPubMed Mudd JC, Brenchley JM. ILC you later: early and irreparable loss of innate lymphocytes in HIV infection. Immunity. 2016;44(2):216–8.CrossRefPubMed
68.
Zurück zum Zitat McKenzie AN, Spits H, Eberl G. Innate lymphoid cells in inflammation and immunity. Immunity. 2014;41(3):366–74.CrossRefPubMed McKenzie AN, Spits H, Eberl G. Innate lymphoid cells in inflammation and immunity. Immunity. 2014;41(3):366–74.CrossRefPubMed
69.
Zurück zum Zitat Krämer B, et al. Compartment-specific distribution of human intestinal innate lymphoid cells is altered in HIV patients under effective therapy. PLoS Pathog. 2017;13(5):e1006373.CrossRefPubMedPubMedCentral Krämer B, et al. Compartment-specific distribution of human intestinal innate lymphoid cells is altered in HIV patients under effective therapy. PLoS Pathog. 2017;13(5):e1006373.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Shelburne SA, et al. Immune reconstitution inflammatory syndrome: emergence of a unique syndrome during highly active antiretroviral therapy. Medicine. 2002;81(3):213–27.CrossRefPubMed Shelburne SA, et al. Immune reconstitution inflammatory syndrome: emergence of a unique syndrome during highly active antiretroviral therapy. Medicine. 2002;81(3):213–27.CrossRefPubMed
71.
Zurück zum Zitat Meya DB, et al. The immunopathogenesis of cryptococcal immune reconstitution inflammatory syndrome: understanding a conundrum. Curr Opin Infect Dis. 2016;29(1):10–22.CrossRefPubMedPubMedCentral Meya DB, et al. The immunopathogenesis of cryptococcal immune reconstitution inflammatory syndrome: understanding a conundrum. Curr Opin Infect Dis. 2016;29(1):10–22.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Wilson EM, Sereti I. Immune restoration after antiretroviral therapy: the pitfalls of hasty or incomplete repairs. Immunol Rev. 2013;254(1):343–54.CrossRefPubMedPubMedCentral Wilson EM, Sereti I. Immune restoration after antiretroviral therapy: the pitfalls of hasty or incomplete repairs. Immunol Rev. 2013;254(1):343–54.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Tran HTT, et al. The role of monocytes in the development of tuberculosis-associated immune reconstitution inflammatory syndrome. Immunobiology. 2014;219(1):37–44.CrossRefPubMed Tran HTT, et al. The role of monocytes in the development of tuberculosis-associated immune reconstitution inflammatory syndrome. Immunobiology. 2014;219(1):37–44.CrossRefPubMed
74.
Zurück zum Zitat Tran HT, et al. Modulation of the complement system in monocytes contributes to tuberculosis-associated immune reconstitution inflammatory syndrome. Aids. 2013;27(11):1725–34.CrossRefPubMed Tran HT, et al. Modulation of the complement system in monocytes contributes to tuberculosis-associated immune reconstitution inflammatory syndrome. Aids. 2013;27(11):1725–34.CrossRefPubMed
75.
Zurück zum Zitat Andrade BB, et al. Mycobacterial antigen driven activation of CD14++ CD16− monocytes is a predictor of tuberculosis-associated immune reconstitution inflammatory syndrome. PLoS Pathog. 2014;10(10):e1004433.CrossRefPubMedPubMedCentral Andrade BB, et al. Mycobacterial antigen driven activation of CD14++ CD16 monocytes is a predictor of tuberculosis-associated immune reconstitution inflammatory syndrome. PLoS Pathog. 2014;10(10):e1004433.CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Pean P, et al. Natural killer cell degranulation capacity predicts early onset of the immune reconstitution inflammatory syndrome (IRIS) in HIV-infected patients with tuberculosis. Blood. 2012;119(14):3315–20.CrossRefPubMed Pean P, et al. Natural killer cell degranulation capacity predicts early onset of the immune reconstitution inflammatory syndrome (IRIS) in HIV-infected patients with tuberculosis. Blood. 2012;119(14):3315–20.CrossRefPubMed
77.
Zurück zum Zitat Conradie F, et al. Natural Killer cell activation distinguishes M. tuberculosis-mediated immune reconstitution syndrome (IRIS) from chronic HIV and HIV-MTB co-infection. J Acquir Immune Defic Syndr (1999). 2011;58(3):309.CrossRef Conradie F, et al. Natural Killer cell activation distinguishes M. tuberculosis-mediated immune reconstitution syndrome (IRIS) from chronic HIV and HIV-MTB co-infection. J Acquir Immune Defic Syndr (1999). 2011;58(3):309.CrossRef
78.
Zurück zum Zitat d’Ettorre G, et al. HIV-associated immune activation: from bench to bedside. AIDS Res Hum Retroviruses. 2011;27(4):355–64.CrossRefPubMed d’Ettorre G, et al. HIV-associated immune activation: from bench to bedside. AIDS Res Hum Retroviruses. 2011;27(4):355–64.CrossRefPubMed
79.
80.
Zurück zum Zitat Appay V, Sauce D. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol. 2008;214(2):231–41.CrossRefPubMed Appay V, Sauce D. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol. 2008;214(2):231–41.CrossRefPubMed
82.
Zurück zum Zitat Kawakami K, Scheidereit C, Roeder RG. Identification and purification of a human immunoglobulin-enhancer-binding protein (NF-kappa B) that activates transcription from a human immunodeficiency virus type 1 promoter in vitro. Proc Natl Acad Sci. 1988;85(13):4700–4.CrossRefPubMedPubMedCentral Kawakami K, Scheidereit C, Roeder RG. Identification and purification of a human immunoglobulin-enhancer-binding protein (NF-kappa B) that activates transcription from a human immunodeficiency virus type 1 promoter in vitro. Proc Natl Acad Sci. 1988;85(13):4700–4.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Appay V, Sauce D. Assessing immune aging in HIV-infected patients. Virulence. 2016;8:1–10. Appay V, Sauce D. Assessing immune aging in HIV-infected patients. Virulence. 2016;8:1–10.
85.
Zurück zum Zitat Hunt PW, Lee SA, Siedner MJ. Immunologic biomarkers, morbidity, and mortality in treated HIV infection. J Infect Dis. 2016;214(Suppl 2):S44–50.CrossRefPubMedPubMedCentral Hunt PW, Lee SA, Siedner MJ. Immunologic biomarkers, morbidity, and mortality in treated HIV infection. J Infect Dis. 2016;214(Suppl 2):S44–50.CrossRefPubMedPubMedCentral
87.
Zurück zum Zitat Ssinabulya I, et al. Subclinical atherosclerosis among HIV-infected adults attending HIV/AIDS care at two large ambulatory HIV clinics in Uganda. PLoS ONE. 2014;9(2):e89537.CrossRefPubMedPubMedCentral Ssinabulya I, et al. Subclinical atherosclerosis among HIV-infected adults attending HIV/AIDS care at two large ambulatory HIV clinics in Uganda. PLoS ONE. 2014;9(2):e89537.CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Nakanjako D, et al. CD4 T-cell activation and reduced regulatory T-cell populations are associated with early development of cataracts among HIV-infected adults in Uganda. Immunol Lett. 2014;161(1):44–9.CrossRefPubMedPubMedCentral Nakanjako D, et al. CD4 T-cell activation and reduced regulatory T-cell populations are associated with early development of cataracts among HIV-infected adults in Uganda. Immunol Lett. 2014;161(1):44–9.CrossRefPubMedPubMedCentral
89.
90.
Zurück zum Zitat Silverberg MJ, Chao C, Abrams DI. New insights into the role of HIV infection on cancer risk. Lancet Oncol. 2009;10(12):1133–4.CrossRefPubMed Silverberg MJ, Chao C, Abrams DI. New insights into the role of HIV infection on cancer risk. Lancet Oncol. 2009;10(12):1133–4.CrossRefPubMed
91.
Zurück zum Zitat Arnsten JH, et al. Decreased bone mineral density and increased fracture risk in aging men with or at risk for HIV infection. AIDS. 2007;21(5):617–23.CrossRefPubMedPubMedCentral Arnsten JH, et al. Decreased bone mineral density and increased fracture risk in aging men with or at risk for HIV infection. AIDS. 2007;21(5):617–23.CrossRefPubMedPubMedCentral
92.
93.
Zurück zum Zitat McCutchan JA, et al. HIV suppression by HAART preserves cognitive function in advanced, immune-reconstituted AIDS patients. AIDS. 2007;21(9):1109–17.CrossRefPubMed McCutchan JA, et al. HIV suppression by HAART preserves cognitive function in advanced, immune-reconstituted AIDS patients. AIDS. 2007;21(9):1109–17.CrossRefPubMed
Metadaten
Titel
Effects of HIV infection and ART on phenotype and function of circulating monocytes, natural killer, and innate lymphoid cells
verfasst von
Rose Nabatanzi
Stephen Cose
Moses Joloba
Sarah Rowland Jones
Damalie Nakanjako
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
AIDS Research and Therapy / Ausgabe 1/2018
Elektronische ISSN: 1742-6405
DOI
https://doi.org/10.1186/s12981-018-0194-y

Weitere Artikel der Ausgabe 1/2018

AIDS Research and Therapy 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.