Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Clin Infect Dis ; 71(6): 1438-1446, 2020 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31608409

RESUMO

BACKGROUND: Cytomegalovirus (CMV) seropositivity and anti-CMV immunoglobulin G (IgG) levels are associated with adverse health outcomes in elderly populations. Among people living with human immunodeficiency virus (PLWH), CMV seropositivity has been associated with persistent CD8 T-cell elevation and increased risk of developing non-AIDS comorbidities despite long-term antiretroviral therapy (ART). Herein, we investigated whether CMV seropositivity and elevation of anti-CMV IgG levels were associated with increased epithelial gut damage, microbial translocation, and systemic inflammation. METHODS: A total of 150 PLWH (79 ART-naive and 71 ART-treated) were compared to 26 without human immunodeficiency virus (HIV) infection (uninfected controls). Plasma markers of HIV disease progression, epithelial gut damage, microbial translocation, nonspecific B-cell activation, anti-CMV and anti-Epstein-Barr virus (EBV) IgG levels, and proinflammatory cytokines were measured. RESULTS: CMV seropositivity and elevated anti-CMV IgG levels were associated with markers of epithelial gut damage, microbial translocation, and inflammation in PLWH and participants without HIV infection. In contrast, total nonspecific IgG, immunoglobulin M, immunoglobulin A, and anti-EBV IgG levels were not associated with these markers. CMV seropositivity was associated with markers of epithelial gut damage, microbial translocation, and inflammation independent of sociodemographic and behavioral characteristics of the study population. CONCLUSIONS: CMV-seropositive people with and without HIV had increased epithelial gut damage, microbial translocation, and inflammation. Furthermore, anti-CMV IgG levels were independently associated with increased epithelial gut damage and microbial translocation. CMV coinfection may partially explain persistent gut damage, microbial translocation, and inflammation in ART-treated PLWH.


Assuntos
Coinfecção , Infecções por Citomegalovirus , Infecções por HIV , Idoso , Anticorpos Antivirais , Citomegalovirus , Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/epidemiologia , HIV , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos
2.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31189714

RESUMO

HIV's ability to persist during suppressive antiretroviral therapy is the main barrier to cure. Immune-privileged tissues, such as the testes, may constitute distinctive sites of HIV persistence, but this has been challenging to study in humans. We analyzed the proviral burden and genetics in the blood and testes of 10 individuals on suppressive therapy who underwent elective gender-affirming surgery. HIV DNA levels in matched blood and testes were quantified by quantitative PCR, and subgenomic proviral sequences (nef region) were characterized from single templates. HIV diversity, compartmentalization, and immune escape burden were assessed using genetic and phylogenetic approaches. Diverse proviruses were recovered from the blood (396 sequences; 354 nef-intact sequences) and testes (326 sequences; 309 nef-intact sequences) of all participants. Notably, the frequency of identical HIV sequences varied markedly between and within individuals. Nevertheless, proviral loads, within-host unique HIV sequence diversity, and the immune escape burden correlated positively between blood and testes. When all intact nef sequences were evaluated, 60% of participants exhibited significant blood-testis genetic compartmentalization, but none did so when the evaluation was restricted to unique sequences per site, suggesting that compartmentalization, when present, is attributable to the clonal expansion of HIV-infected cells. Our observations confirm the testes as a site of HIV persistence and suggest that individuals with larger and more diverse blood reservoirs will have larger and more diverse testis reservoirs. Furthermore, while the testis microenvironment may not be sufficiently unique to facilitate the seeding of unique viral populations therein, differential clonal expansion dynamics may be at play, which may complicate HIV eradication.IMPORTANCE Two key questions in HIV reservoir biology are whether immune-privileged tissues, such as the testes, harbor distinctive proviral populations during suppressive therapy and, if so, by what mechanism. While our results indicated that blood-testis HIV genetic compartmentalization was reasonably common (60%), it was always attributable to differential frequencies of identical HIV sequences between sites. No blood-tissue data set retained evidence of compartmentalization when only unique HIV sequences per site were considered; moreover, HIV immune escape mutation burdens were highly concordant between sites. We conclude that the principal mechanism by which blood and testis reservoirs differ is not via seeding of divergent HIV sequences therein but, rather, via differential clonal expansion of latently infected cells. Thus, while viral diversity and escape-related barriers to HIV eradication are of a broadly similar magnitude across the blood and testes, clonal expansion represents a challenge. The results support individualized analysis of within-host reservoir diversity to inform curative approaches.


Assuntos
Antirretrovirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , HIV-1/classificação , Testículo/virologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Estudos de Casos e Controles , Evolução Clonal , Procedimentos Cirúrgicos Eletivos , Variação Genética , Infecções por HIV/sangue , HIV-1/efeitos dos fármacos , HIV-1/genética , Humanos , Masculino , Filogenia , Análise de Sequência de RNA , Cirurgia de Readequação Sexual , Testículo/efeitos dos fármacos , Testículo/cirurgia
3.
BMJ Open ; 9(4): e028444, 2019 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-31005944

RESUMO

INTRODUCTION: People living with HIV (PLWH) on antiretroviral therapy (ART) do not progress to AIDS. However, they still suffer from an increased risk of inflammation-associated complications. HIV persists in long-lived CD4+ T cells, which form the major viral reservoir. The persistence of this reservoir despite long-term ART is the major hurdle to curing HIV. Importantly, the size of the HIV reservoir is larger in individuals who start ART late in the course of infection and have a low CD4+/CD8+ ratio. HIV reservoir size is also linked to the levels of persistent inflammation on ART. Thus, novel strategies to reduce immune inflammation and improve the host response to control the HIV reservoir would be a valuable addition to current ART. Among the different strategies under investigation is metformin, a widely used antidiabetic drug that was recently shown to modulate T-cell activation and inflammation. Treatment of non-diabetic individuals with metformin controls inflammation by improving glucose metabolism and by regulating intracellular immunometabolic checkpoints such as the adenosin 5 monophosphate activated protein kinase and mammalian target of rapamycin, in association with microbiota modification. METHODS AND ANALYSIS: 22 PLWH on ART for more than 3 years, at high risk of inflammation or the development of non-AIDS events (low CD4+/CD8+ ratio) will be recruited in a clinical single-arm pilot study. We will test whether supplementing ART with metformin in non-diabetic HIV-infected individuals can reduce the size of the HIV reservoir as determined by various virological assays. The expected outcome of this study is a reduction in both the size of the HIV reservoir and inflammation following the addition of metformin to ART, thus paving the way towards HIV eradication. ETHICS AND DISSEMINATION: Ethical approval: McGill university Health Centre committee number MP-37-2016-2456. Canadian Canadian Institutes of Health Research/Canadian HIV Trials Network (CTN) protocol CTNPT027. Results will be made available through publication in peer-reviewed journals and through the CTN website. TRIAL REGISTRATION NUMBER: NCT02659306.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Reservatórios de Doenças/virologia , Infecções por HIV/tratamento farmacológico , Metformina/uso terapêutico , Adulto , Feminino , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Carga Viral , Adulto Jovem
4.
PLoS One ; 13(3): e0194185, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29601578

RESUMO

Interleukin-18 (IL-18) is a pleiotropic cytokine of the IL-1 family with multiple context dependent functions. We and others have shown that HIV infection is accompanied by increased circulating levels of IL-18 along with decreased levels of its antagonist, Interleukin-18 Binding Protein (IL-18BP). The infection is also accompanied by intestinal inflammation and decreased intestinal integrity as measured by intestinal permeability, regeneration and repair. However, little is known concerning the relation between high level of IL-18 associated with the viral infection and intestinal permeability. Here we demonstrate that HIV treatment increases production of IL-18 and decreases that of IL-18BP production in human intestinal epithelial cell (IEC) lines. IL-18 causes apoptosis of the IEC by activating caspase-1 and caspase-3. It induces epithelial barrier hyperpermeability by decreasing and disrupting both tight and adherens junction proteins, occludin, claudin 2 and beta-catenin. Disorganization of F-actin was also observed in the IEC that were exposed to the cytokine. Moreover IL-18 decreases transepithelial electrical resistance (TEER) in Caco-2 and increases permeability in HT29 monolayers. The cells' treatment with IL-18 causes an increase in the expression of phosphorylated myosin II regulatory light-chain (p-MLC) and myosin light-chain kinase (MLCK), and a decrease in phosphorylated Signal Transducer and Activator of Transcription (p-STAT)-5. This increase in p-MLC is suppressed by a Rho-kinase (ROCK)-specific inhibitor. Interestingly, the levels of the cytokine correlate with those of LPS in the circulation in three different categories of HIV infected patients (HAART-naïve and HAART-treated HIV-infected individuals, and Elite controls) as well as in healthy controls. Collectively, these results suggest that the HIV-induced IL-18 plays a role in increased intestinal permeability and microbial translocation observed in HIV-infected individuals.


Assuntos
Translocação Bacteriana , Células Epiteliais/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Interleucina-18/biossíntese , Mucosa Intestinal/metabolismo , Células CACO-2 , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Infecções por HIV/microbiologia , Infecções por HIV/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Masculino , Permeabilidade
5.
J Reprod Immunol ; 125: 16-24, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29136520

RESUMO

The testis has been described in animal models as a site of immune privilege, which protects spermatids against tissue damage during inflammation. Myeloid cells, including macrophages and dendritic cells (DC), are defined as key players in the testicular immune privilege in animal models. However, their distribution and frequency in human testis remain poorly described. To overcome the challenges related to tissue sampling, we obtained testicular tissue from men under hormonal therapy who elected to have sex reassignment surgery (SRS). We examined the distribution of myeloid cell populations in tissue sections using immunohistofluorescence and evaluated their relative frequencies in fresh testicular cell suspensions compared with matched blood using multi-parametric flow cytometry. We identified 4.9% of CD45+ leucocytes in testicular cell suspensions, of which 0.4% were B cells, demonstrating a low level of blood contamination. Myeloid cells (Lin-HLA-DR+) were located in the testicular interstitium and represented a median of 23.4% of testicular leucocytes, displaying higher HLA-DR expression compared to their counterparts in blood (p=0.001). The frequency of testicular myeloid cells was not linked with the duration of hormonal therapy. Resident macrophages (CD14+CD163+) constituted the most frequent myeloid cell subset and expressed high levels of CD163. Elevated proportion of myeloid DC (CD14-CD11c+) contrasted with the paucity of plasmacytoïd DC (CD14-CD123+) in testis. Myeloid-derived suppressor cells (Lin-HLA-DR-CD33hiCD11bhi) were not detected in the testis while constituting 0.5% of blood leucocytes. For the first time, we characterized myeloid cell subsets in human testes collected after SRS, providing a basis to assess their contribution to immune privilege.


Assuntos
Células Dendríticas/imunologia , Terapia de Reposição Hormonal/métodos , Macrófagos/imunologia , Células Supressoras Mieloides/imunologia , Testículo/citologia , Adulto , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/imunologia , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antígeno CD11b/imunologia , Antígeno CD11b/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Estradiol/administração & dosagem , Humanos , Subunidade alfa de Receptor de Interleucina-3/imunologia , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Receptores de Lipopolissacarídeos/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Células Supressoras Mieloides/efeitos dos fármacos , Células Supressoras Mieloides/metabolismo , Orquiectomia , Progesterona/administração & dosagem , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Cirurgia de Readequação Sexual , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Espironolactona/administração & dosagem , Testículo/efeitos dos fármacos , Testículo/imunologia , Testículo/metabolismo
6.
J Virus Erad ; 3(4): 212-217, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-29057085

RESUMO

INTRODUCTION: Antiretroviral therapy (ART) does not cure HIV infection due to the persistence of HIV reservoirs in long-lived memory CD4 T cells present in the blood, lymph nodes, intestinal tract, and other tissues. Interest grows in obtaining gut-tissue samples for HIV persistence studies, which poses an ethical challenge to provide study volunteers with adequate information on risks and benefits. Herein we assess the risks and benefits of undergoing gut biopsy procedures for HIV pathogenesis and reservoir studies. METHODS: A group discussion was organised with physicians and community representatives on performing either a flexible sigmoidoscopy or a colonoscopy. Consensus was reached on conducting colonoscopy in persons ≥50 years. Thirty HIV-infected, ART-treated and nine uninfected participants were recruited. Colonoscopy was performed to collect 30 gut mucosal biopsies. When present, polyps were removed and abnormal mucosal findings were biopsied for pathological analysis. Participants were interviewed on potential discomfort following colonoscopic examination. RESULTS: The HIV-infected and uninfected groups were comparable in terms of age and gender with more men who have sex with men (MSM) in the former group. Abnormal colonoscopic findings were observed in 43.6% of all the participants and did not differ by HIV status. In total, 24 polyps were removed with a higher mean number of polyps removed in HIV-infected versus uninfected participants (1.7 vs 1.0, P=0.013). The number of polyps marginally correlated with inverted CD4:CD8 ratio. Based on our findings, colonoscopic examination was safe to use for gut biopsy procedures where almost half of the participants had polyps removed. CONCLUSION: Participation in the study provided colon cancer screening as an ancillary benefit that participants could have received in standard medical care, thus mitigating burdens of invasive procedures. Dialogue between community representatives and clinical researchers can increase participation and advance HIV cure research.

7.
Open AIDS J ; 11: 32-35, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28553430

RESUMO

INTRODUCTION: Interleukin-33 (IL-33) is a cell damage-induced alarmin. The plasma concentration of suppression of tumorogenicity (sST2), a surrogate marker of IL-33 production, is a prognostic marker of cardiovascular disease. OBSERVATION: Recently, we reported that sST2 plasma levels were elevated in early HIV-1 infection and linked to markers of microbial translocation and of T cell activation. RESULTS: Here we show that it is not the case in patients with suppressed viremia. Thus, IL-33 plays its alarmin role only during the early phase of the infection.

8.
Oncoimmunology ; 6(1): e1258506, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28197380

RESUMO

The intestinal microbiota plays a key role in the pathogenesis of acute graft-versus-host disease (aGVHD). High-dose conditioning regimens given prior to allogeneic hematopoietic stem cell transplantation (aHSCT) modulate the composition of gut microbiota and damage the gut epithelial barrier, resulting in increased systemic inflammation. We assessed whether gut decontamination with antibiotics (ATB) prior to aHSCT influenced the frequency of aGVHD and mortality in 500 patients from two Canadian centers between 2005 and 2012. The rate of grade II-IV aGVHD was higher in the ATB arm compared with the arm without ATB (42% vs 28%; p < 0.001). This difference was mainly driven by a 2-fold higher rate of grade II-IV gastrointestinal aGVHD (GI-GVHD) in the ATB arm compared with the arm without ATB (20.7% vs 10.8%; p = 0.003). Multivariate analyses adjusted for known aGVHD risk factors revealed that more patients in the ATB group developed clinically significant GI-GVHD and liver aGVHD; adjusted odds ratio (aOR) = 1.83; p = 0.023 and aOR = 3.56; p = 0.047, respectively. Importantly, median overall survival (OS) was significantly lower in the group receiving ATB and the OS at 10 y remained decreased in the ATB group; adjusted hazard ratio (aHR) = 1.61 (p < 0.001). Without undermining the role of ATB prophylaxis to prevent infection in aHSCT, we have shown that the use of ATB that targets intestinal bacteria is associated with a more severe aGVHD that involves the GI organs and impacts OS. Prospective studies that evaluate the contribution of bacterial decontamination to aGVHD are warranted.

9.
Int J Tryptophan Res ; 9: 67-77, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27773992

RESUMO

The term "immune privilege" was originally coined to describe the suppression of inflammatory responses within organs protected by anatomic barriers, ie, the eyes, brain, placenta, and testes. However, cellular and metabolic processes, which orchestrate immune responses, also control inflammation within these sites. Our current understanding of tolerogenic mechanisms has extended the definition of immune privilege to include hair follicles, the colon, and cancer. By catabolizing tryptophan, cells expressing the enzyme indoleamine-2,3-dioxygenase produce kynurenine metabolites, which orchestrate local and systemic responses to control inflammation, thus maintaining immune privilege. This review highlights the double-edged role played by the kynurenine pathway (KP), which establishes and maintains immune-privileged sites while contributing to cancer immune escape. The identification of the underlying molecular drivers of the KP in immune-privileged sites and in cancer is essential for the development of novel therapies to treat autoimmunity and cancer and to improve transplantation outcomes.

10.
AIDS ; 30(18): 2777-2786, 2016 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-27677162

RESUMO

OBJECTIVE: HIV persistence in long-lived infected cells and in anatomical sanctuary sites are major hurdles to HIV eradication. Testicular tissue may represent a significant viral sanctuary site as it constitutes an immunologically privileged compartment. We assessed immunotolerance properties of the testicular tissue in individuals receiving suppressive antiretroviral therapy (ART). DESIGN AND METHODS: Testicular tissue and matched blood samples were collected from six virally suppressed adults and 10 HIV-uninfected controls prior to sex reassignment surgery. T cells were purified from freshly isolated testicular interstitial cell suspensions. T-cell subsets, expression of immune activation markers and HIV DNA were assessed in matched testicular cells and peripheral blood mononuclear cells (PBMCs). RESULTS: When compared with PBMCs, testes were characterized by a lower CD4 T-cell proportion among total T cells, a decrease in the frequency of naive cells, an increase in the frequency of effector-memory T cells and an increase in CCR5 expression in both the HIV+ and HIV- groups. In HIV-infected individuals on ART, testes displayed higher T-cell immune activation (Coexpression of CD38 and Human Leukocyte Antigen - antigen D Related) than PBMCs. In both groups, testes were characterized by higher frequencies of immunosuppressive CD39 regulatory T cells and a massive increase in CD73 expression on CD8 T cells. In addition, a remarkable increase in indoleamine-pyrrole 2,3-dioxygenase immunosuppressive enzyme involved in tryptophan/kynurenine catabolism was observed in testes versus blood. Rare cells harboring HIV DNA were detected in testes from five out six participants. CONCLUSION: These findings suggest that the adenosine and tryptophan/kynurenine immune-metabolic pathways contribute to immune tolerance in testicular tissue. Our results suggest that testes may represent a distinctive HIV sanctuary site during ART.


Assuntos
Antirretrovirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Tolerância Imunológica , Subpopulações de Linfócitos T/imunologia , Testículo/imunologia , Testículo/virologia , Adulto , DNA Viral/análise , Perfilação da Expressão Gênica , Humanos , Fatores Imunológicos/análise , Masculino
11.
EBioMedicine ; 9: 37-44, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27397514

RESUMO

UNLABELLED: Interleukin 33 (IL-33), a member of the IL-1 family, is constitutively expressed in epithelial and in endothelial cells at barrier sites, acting as a danger signal and adjuvanting the immune response following tissue damage and infection. Originally implicated in allergy, IL-33 is also known to be involved in innate and adaptive immune responses by enhancing natural killer, Th1, and CD4 and CD8 T-cell functions. The nature of the antiviral immune response orchestrated by IL-33 depends on the site of infection, the duration of the disease and the cytokine milieu. In this review, we focus on the distinctive contribution of IL-33 as an anti-infective and proinflammatory cytokine in response to cell death and viral infections. The dynamic role of IL-33 in the acute and chronic phases of infection with HIV, hepatitis B and C viruses, and with CMV is highlighted. This review will also discuss the potential immunotherapeutic and adjuvant roles of IL-33. SEARCH STRATEGY AND SELECTION CRITERIA: English language, indexed publications in PubMed were searched using combinations of following key words: "interleukin-33", "IL-33", "suppression of tumorigenicity 2", ST2", "sST2", "HIV", "HBV", "HCV", "CMV", "HPV", "immunotherapy" and "vaccine". Except for seminal studies, only articles published between 2010 and 2016 were included.


Assuntos
Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Transdução de Sinais , Viroses/metabolismo , Viroses/virologia , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Coinfecção , Humanos , Transdução de Sinais/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Tacrolimo/análogos & derivados , Viroses/tratamento farmacológico , Viroses/imunologia
12.
AIDS ; 30(10): 1617-27, 2016 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-27045377

RESUMO

OBJECTIVE: Following tissue barrier breaches, interleukin-33 (IL-33) is released as an 'alarmin' to induce inflammation. Soluble suppression of tumorigenicity 2 (sST2), as an IL-33 decoy receptor, contributes to limit inflammation. We assessed the relationship between the IL-33/ST2 axis and markers of gut mucosal damage in patients with early (EHI) and chronic HIV infection (CHI) and elite controllers. DESIGN: Analyses on patients with EHI and CHI were conducted to determine IL-33/sST2 changes over time. METHODS: IL-33 and sST2 levels were measured in plasma. Correlations between sST2 levels and plasma viral load, CD4 and CD8 T-cell counts, expression of T-cell activation/exhaustion markers, gut mucosal damage, microbial translocation and inflammation markers, as well as kynurenine/tryptophan ratio were assessed. RESULTS: Plasma sST2 levels were elevated in EHI compared with untreated CHI and uninfected controls, whereas IL-33 levels were comparable in all groups. In EHI, sST2 levels were positively correlated with the CD8 T-cell count and the percentage of T cells expressing activation and exhaustion markers, but not with viral load or CD4 T-cell count. Plasma sST2 levels also correlated with plasma levels of gut mucosal damage, microbial translocation and kynurenine/tryptophan ratio and for some markers of inflammation. Prospective analyses showed that early antiretroviral therapy had no impact on sST2 levels, whereas longer treatment duration initiated during CHI normalized sST2. CONCLUSION: As sST2 levels were elevated in EHI and were correlated with CD8 T-cell count, immune activation, and microbial translocation, sST2 may serve as a marker of disease progression, gut damage and may directly contribute to HIV pathogenesis.


Assuntos
Infecções por HIV/patologia , Proteína 1 Semelhante a Receptor de Interleucina-1/sangue , Mucosa Intestinal/patologia , Plasma/química , Adulto , Translocação Bacteriana , Estudos Transversais , Feminino , Humanos , Interleucina-33/sangue , Ativação Linfocitária , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Linfócitos T/química , Linfócitos T/imunologia , Carga Viral , Adulto Jovem
13.
Cytokine Growth Factor Rev ; 28: 1-10, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26851985

RESUMO

HIV-1 infection leads to a depletion of CD4 T-cells associated with a persistent immune inflammation and changes in cellular metabolism. Most effort of managing HIV infection with combination of antiretroviral therapies (ART) has been focused on CD4 T-cell recovery, while control of persistent immune inflammation and metabolism were relatively underappreciated in the past. Recent discoveries on the interplay between innate immunity, inflammation (especially the inflammasome) and metabolic changes in the context of cancer and autoimmunity provide an emerging field for chronic viral infections including HIV-1. In a previous review, we described the deregulated metabolism contributing to immune dysfunctions such as alteration of memory T-cell responses, mucosal protection, and dendritic cell-related antigen presentation. Here, we summarize the latest knowledge on the detrimental influence of long-lasting inflammation and inflammasome activation induced by HIV-1, gut dysbiosis, and bacterial translocation, on metabolism during the course of viral infection. We also report on the inability of ART to fully counteract inflammation, resulting in partial metabolic improvement and leading to an insufficient decrease in the risk of non-AIDS events. Further advances in our understanding of the relationship between inflammation, altered metabolism, and long-term ART is warranted. Additionally, there is a critical need for developing new strategies to regulate the pro-inflammatory signals to enhance cellular metabolism and immune functions in order to improve the quality of life of individuals living with HIV-1.


Assuntos
Infecções por HIV/metabolismo , HIV-1/imunologia , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , Infecções por HIV/patologia , Infecções por HIV/virologia , Humanos , Imunidade Inata/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/virologia , Qualidade de Vida
14.
Expert Rev Clin Immunol ; 12(2): 91-4, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26629806

RESUMO

Antiretroviral therapy (ART) has reshaped the lives of millions of individuals infected with human immunodeficiency virus (HIV). Patients initiating ART early in the course of infection benefit from a considerable reduction in the risks of acquired immune deficiency syndrome (AIDS) and HIV-related inflammatory events. However, the absence of cure and lifelong requirements of treatment highlight the need of a vaccine and an immunotherapeutic strategy. Like for cancer, a paradigm shift has occurred with the contribution of immune activation and microbial translocation priming aberrant systemic immunity in restricting the ability of the host to mount an effective immune response. The approaches of implementing an effective vaccine to prevent infection and inhibition of immune activation with breakage of viral latency followed by vaccination should lead to an HIV-free generation.


Assuntos
Vacinas contra a AIDS/imunologia , Síndrome da Imunodeficiência Adquirida/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Imunoterapia/métodos , Vacinas contra a AIDS/uso terapêutico , Síndrome da Imunodeficiência Adquirida/prevenção & controle , Síndrome da Imunodeficiência Adquirida/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Previsões , Infecções por HIV/prevenção & controle , Infecções por HIV/terapia , HIV-1/fisiologia , Humanos , Imunoterapia/tendências , Ativação Linfocitária/imunologia , Vacinação/métodos , Vacinação/tendências
15.
AIDS Rev ; 17(2): 96-106, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26035167

RESUMO

Tryptophan degradation along the kynurenine pathway is associated with a wide variety of pathophysiological processes, of which tumor tolerance and immune dysfunction in several chronic viral infections including HIV are well known. The kynurenine pathway is at the crossroads of metabolism and immunity and plays an important role in inflammation while also playing an opposing role in the control of acute and chronic infections. In this review we have summarized findings from recent studies reporting modulation of tryptophan degrading the kynurenine pathway in the context of HIV infection. This immuno-metabolic pathway is modulated by three distinct inducible enzymes: indoleamine 2,3-dioxygenase 1 and 2 and tryptophan 2,3-dioxygenase. Increased expression of these enzymes by antigen-presenting cells leads to local or systemic tryptophan depletion, resulting in a mechanism of defense against certain microorganisms. Conversely, it can also lead to immunosuppression by antigen-specific T-cell exhaustion and recruitment of T regulatory cells. Recently, among these enzymes, indoleamine 2,3-dioxygenase 1 has been recognized to be an immune response checkpoint that plays an important role in HIV immune dysfunction, even in the context of antiretroviral therapy. In addition to the activation of the kynurenine pathway by HIV proteins Tat and Nef, the tryptophan-degrading bacteria present in the intestinal flora have been associated with dysfunction of gut mucosal CD4 Th17/Th22 cells, leading to microbial translocation and creating a systemic kynurenine pathway activation cycle. This self-sustaining feedback loop has deleterious effects on disease progression and on neurocognitive impairment in HIV-infected patients. Therapy designed to break the vicious cycle of induced tryptophan degradation is warranted to revert immune exhaustion in HIV-infected persons.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/patologia , Inflamação , Cinurenina/metabolismo , Redes e Vias Metabólicas , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Bactérias/metabolismo , Translocação Bacteriana , Interações Hospedeiro-Patógeno , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Triptofano/metabolismo , Triptofano Oxigenase/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
16.
J Trauma Acute Care Surg ; 76(1): 241-4, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24368387

RESUMO

BACKGROUND: Immune system biology and most physiologic functions are tightly linked to circadian rhythms. Time of day-dependent variations in many biologic parameters also play a fundamental role in the disease process. We previously showed that the genes encoding the peripheral molecular clock were modulated in a sex-dependent manner in Q fever. METHODS: Here, we examined severe trauma patients at admission to the intensive care unit. Using quantitative real-time polymerase chain reaction, the whole-blood expression of the molecular clock components ARNTL, CLOCK, and PER2 was assessed in male and female trauma patients. Healthy volunteers of both sexes were used as controls. RESULTS: We observed a significant overexpression of both ARNTL and CLOCK in male trauma patients. CONCLUSION: We report, for the first time, the sex-related modulation of the molecular clock genes in the blood following severe trauma. These results emphasize the role of circadian rhythms in the immune response in trauma patients. LEVEL OF EVIDENCE: Epidemiologic study, level IV.


Assuntos
Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/sangue , Ferimentos e Lesões/fisiopatologia , Fatores de Transcrição ARNTL/sangue , Adulto , Proteínas CLOCK/sangue , Estudos de Casos e Controles , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Circadianas Period/sangue , Reação em Cadeia da Polimerase em Tempo Real , Fatores Sexuais , Ferimentos e Lesões/sangue
17.
J Infect Dis ; 208(6): 942-51, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23801603

RESUMO

BACKGROUND: Q fever is caused by Coxiella burnetii, a bacterium that persists in M2-polarized macrophages. We wondered whether the concept of M1/M2 polarization is applicable to Q fever patients. METHODS: Monocytes from healthy controls were cultured with IFN-γ and IL-4, agonists of M1 and M2 macrophages, respectively, and their gene expression was assessed using whole-genome microarrays. Selected biomarkers were assessed in blood from Q fever patients by real-time reverse transcription polymerase chain reaction (RT-PCR). RESULTS: Monocytes exhibited early (6-hour) patterns of activation specific to IFN-γ or IL-4 and a late (18-hour) pattern of common activation. Because these responses were not reducible to M1/M2 polarization, we selected biomarkers and tested their relevance in Q fever patients. The early genes NLRC5, RTP4, and RHOH, which were modulated in response to IFN-γ, were up-regulated in patients with acute Q fever, and the expression levels of the late genes ALOX15, CLECSF1, CCL13, and CCL23 were specifically increased in patients with Q fever endocarditis. The RHOH and ALOX15 genes were associated with the activity of acute Q fever and Q fever endocarditis, respectively. CONCLUSIONS: Our results show that the kinetic model of monocyte activation enables a dynamic approach for the evaluation of Q fever patients.


Assuntos
Ativação de Macrófagos , Monócitos/imunologia , Febre Q/imunologia , Doença Aguda , Adulto , Idoso , Biomarcadores/sangue , Estudos de Casos e Controles , Coxiella burnetii , Endocardite Bacteriana/imunologia , Regulação da Expressão Gênica , Humanos , Interferon gama/imunologia , Interleucina-4/imunologia , Macrófagos/imunologia , Análise em Microsséries , Pessoa de Meia-Idade , Transcrição Gênica , Regulação para Cima
18.
Eur J Cell Biol ; 91(11-12): 938-49, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22999511

RESUMO

Leukocytes migrate through most tissues in the body, a process which takes place in 3D environments. We have previously shown that macrophages use the amoeboid migration mode in porous matrices such as fibrillar collagen I and the mesenchymal mode involving podosomes and matrix proteolysis in dense matrices such as Matrigel. Whether such a plasticity may apply to other leukocytes and to all subsets of macrophages is unknown. Here, we therefore provide a comparative analysis of the in vitro 3D migration modes adopted by primary human leukocytes. Blood-derived monocytes, neutrophils and T lymphocytes were found to use the amoeboid mode in a porous fibrillar collagen I matrix but were unable to infiltrate dense Matrigel and to form podosomes. M2-polarized macrophages and elicited peritoneal macrophages formed podosome rosettes, degraded the ECM and infiltrated both matrices. In contrast, M1 macrophages were motionless in 2D and 3D environments, whilst resident macrophages, devoid of podosomes, were only able to use the amoeboid mode. Thus, we conclude that whereas all leukocytes use the amoeboid mode to migrate through porous matrices, it is only certain macrophages that can adopt the mesenchymal mode that permits migration through dense matrices. Interestingly, the acquisition of mesenchymal migration capacity by macrophages correlates with the presence of podosomes and with their capacity to organize those as rosettes, which appears to be modulated by their differentiation and polarization states. As a perspective, specific control of the mesenchymal migration would be a potential target for therapeutic approaches aiming at decreasing macrophage tissue infiltration.


Assuntos
Movimento Celular , Extensões da Superfície Celular/fisiologia , Leucócitos/fisiologia , Leucócitos/ultraestrutura , Macrófagos/fisiologia , Macrófagos/ultraestrutura , Extensões da Superfície Celular/ultraestrutura , Colágeno , Colágeno Tipo I/química , Combinação de Medicamentos , Matriz Extracelular/química , Humanos , Laminina , Conformação Molecular , Fenótipo , Proteoglicanas
19.
FEMS Immunol Med Microbiol ; 64(1): 98-100, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22066909

RESUMO

Q fever is a disease caused by Coxiella burnetii, an obligate intracellular bacterium. Acute Q fever is spontaneously resolutive and is characterized by an efficient immune response. In contrast, chronic Q fever is characterized by dysregulated immune response, as demonstrated by the failure of C. burnetii to induce lymphoproliferation and the lack of granulomas. Recently, it has been demonstrated that when co-expressed in heterologous mammalian cell lines, the ligands of Numb proteins X1 and X2 (LNX1 and LNX2) regulate the level of the T-cell co-receptor CD8, which plays an essential role in T-cell-mediated immune response. We decided to investigate the expression of LNX1 and LNX2 genes in patients with acute or chronic Q fever. Interestingly, we found a high level of LNX1 and LNX2 mRNAs in endocarditis, the principal manifestation of chronic Q fever, but not in acute Q fever. Our data suggest that LNXs may be used as complementary biomarkers to follow the prognosis of chronic Q fever.


Assuntos
Proteínas de Transporte/biossíntese , Coxiella burnetii/patogenicidade , Febre Q/diagnóstico , Febre Q/patologia , Ubiquitina-Proteína Ligases/biossíntese , Biomarcadores , Linfócitos T CD8-Positivos/imunologia , Perfilação da Expressão Gênica , Humanos , Prognóstico , RNA Mensageiro/biossíntese
20.
Curr Opin Infect Dis ; 24(3): 230-4, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21311324

RESUMO

PURPOSE OF REVIEW: Macrophages are the first line of defense against pathogens, and the mode of their activation will determine the success or failure of the host response to pathogen aggression. Based on limited numbers of markers, activated macrophages can be classified as classically activated (M1) macrophages that support microbicidal activity or alternatively activated (M2) macrophages that are not competent to eliminate pathogens. The development of high-throughput gene expression methods affords a reappraisal of the concept of macrophage activation in human infectious diseases. RECENT FINDINGS: By combining microarray data and conventional approaches, it is becoming clear that the M1 polarization program is associated with gastrointestinal infections (e.g. typhoid fever and Helicobacter pylori gastritis) and active tuberculosis. An M2 signature is observed in lepromatous leprosy, Whipple's disease, and localized infections (keratitis, chronic rhinosinusitis). However, these findings could not be predicted from the analysis of the M1/M2 programs of macrophages stimulated in vitro. SUMMARY: The reappraisal of macrophage polarization by high-throughput methods is critical to understanding the role of macrophage polarization in infectious diseases. Only the identification of individual profiles will support promising therapeutic approaches based on target determination.


Assuntos
Infecções Bacterianas/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Perfilação da Expressão Gênica , Humanos , Análise em Microsséries
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA