Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
PLOS Glob Public Health ; 3(12): e0002339, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38117812

RESUMEN

Diversity, equity and inclusion (DEI) in science is vital to improve the scientific process and ensure societal uptake and application of scientific results. DEI challenges include a full spectrum of issues from the lack of, and promotion of, women in science, to the numerous barriers in place that limit representation of African scientists in global scientific efforts. DEI principles in African science remain relatively underdeveloped, with limited engagement and discussion among all stakeholders to ensure that initiatives are relevant to local environments. The Sub-Saharan African Network for TB/HIV research Excellence (SANTHE) is a network of African-led research in HIV, tuberculosis (TB), associated co-morbidities, and emerging pathogens, now based in eight African countries. Our aim, as a scientific capacity strengthening network, was to collaboratively produce a set of DEI guidelines and to represent them visually as a DEI compass. We implemented a consortium-wide survey, focus group discussions and a workshop where we were able to identify the key DEI challenges as viewed by scientists and support staff within the SANTHE network. Three thematic areas were identified: 1. Conquering Biases, 2. Respecting the Needs of a Diverse Workforce (including mental health challenges, physical disability, career stability issues, demands of parenthood, and female-specific challenges), and 3. Promotion of African Science. From this we constructed a compass that included proposed steps to start addressing these issues. The use of the compass metaphor allows 're-adjustment/re-positioning' making this a dynamic output. The compass can become a tool to establish an institution's DEI priorities and then to progress towards them.

2.
Health Res Policy Syst ; 21(1): 109, 2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880720

RESUMEN

Recent efforts to shift the control and leadership of health research on African issues to Africa have led to increased investments for scientific research capacity strengthening (RCS) on the continent and a greater demand for accountability, value for money and demonstration of return on investment. There is limited literature on monitoring and evaluation (M&E) of RCS systems and there is a clear need to further explore whether the M&E frameworks and approaches that are currently used are fit for purpose. The M&E approaches taken by four African RCS consortia funded under the Developing Excellence in Leadership, Training and Science in Africa (DELTAS) I initiative were assessed using several methods, including a framework comparison of the M&E approaches, semi-structured interviews and facilitated discussion sessions. The findings revealed a wide range in the number of indicators used in the M&E plans of individual consortium, which were uniformly quantitative and at the output and outcome levels. Consortia revealed that additional information could have been captured to better evaluate the success of activities and measure the ripple effects of their efforts. While it is beneficial for RCS consortia to develop and implement their own M&E plans, this could be strengthened by routine engagement with funders/programme managers to further align efforts. It is also important for M&E plans to consider qualitative data capture for assessment of RCS efforts. Efforts could be further enhanced by supporting platforms for cross-consortia sharing, particularly when trying to assess more complex effects. Consortia should make sure that processes for developmental evaluation, and capturing and using the associated learning, are in place. Sharing the learning associated with M&E of RCS efforts is vital to improve future efforts. Investing and improving this aspect of RCS will help ensure tracking of progress and impact of future efforts, and ensure accountability and the return on investment. The findings are also likely applicable well beyond health research.


Asunto(s)
Creación de Capacidad , Inversiones en Salud , Humanos , África , Exactitud de los Datos
3.
EBioMedicine ; 75: 103787, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34968761

RESUMEN

BACKGROUND: Biomarkers correlating with Mycobacterium tuberculosis infection activity/burden in asymptomatic individuals are urgently needed to identify and treat those at highest risk for developing active tuberculosis (TB). Our main objective was to identify plasma host protein biomarkers that change over time prior to developing TB in people living with HIV (PLHIV). METHODS: Using multiplex MRM-MS, we investigated host protein expressions from 2 years before until time of TB diagnosis in longitudinally collected (every 3-6 months) and stored plasma from PLHIV with incident TB, identified within a South African (SA) and US cohort. We performed temporal trend and discriminant analyses for proteins, and, to assure clinical relevance, we further compared protein levels at TB diagnosis to interferon-gamma release assay (IGRA; SA) or tuberculin-skin test (TST; US) positive and negative cohort subjects without TB. SA and US exploratory data were analyzed separately. FINDINGS: We identified 15 proteins in the SA (n=30) and 10 in the US (n=24) incident TB subjects which both changed from 2 years prior until time of TB diagnosis after controlling for 10% false discovery rate, and were significantly different at time of TB diagnosis compared to non-TB subjects (p<0.01). Five proteins, CD14, A2GL, NID1, SCTM1, and A1AG1, overlapped between both cohorts. Furthermore, after cross-validation, panels of 5 - 12 proteins were able to predict TB up to two years before diagnosis. INTERPRETATION: Host proteins can be biomarkers for increasing Mycobacterium tuberculosis infection activity/burden, incipient TB, and predict TB development in PLHIV. FUNDING: NIH/NIAID AI117927, AI146329, and AI127173 to JMA.


Asunto(s)
Infecciones por VIH , Mycobacterium tuberculosis , Tuberculosis , Biomarcadores , Infecciones por VIH/complicaciones , Humanos , Prueba de Tuberculina , Tuberculosis/complicaciones , Tuberculosis/diagnóstico
4.
Wellcome Open Res ; 7: 87, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37456907

RESUMEN

Underrepresentation of women in scientific leadership is a global problem. To understand and counter narratives that limit gender equity in African science, we conducted a public engagement campaign. Scientists representing six sub-Saharan African countries and multiple career stages used superhero imagery to create a diverse and unified team advocating for gender equity in science. In contrast to many traditional scientific environments and global campaigns, this "PowerPack of SuperScientists" was led by early-career Black female scientists whose perspectives are often under-represented in discussions about gender equity in science. The superhero imagery served as a powerful and fun antidote to imposter syndrome and helped to subvert traditional power structures based on age, race and sex. In an interactive social media campaign, the PowerPack developed insights into three themes: a) cultural stereotypes that limit women's scientific careers, b) the perception of a "conflict" between family and career responsibilities for women scientists, and c) solutions that can be adopted by key stakeholders to promote gender equity in African science. The PowerPack proposed solutions that could be undertaken by women working individually or collectively and interventions that require allyship from men, commitment from scientific institutions, and wider societal change. Further work is required to fully engage African scientists from even more diverse and disadvantaged backgrounds and institutions in these solutions and to enhance commitment by different stakeholders to achieving gender equity in science. Our experience suggests that creative tools should be used to subvert power dynamics and bring fresh perspectives and urgency to this topic.

5.
BMC Public Health ; 20(1): 1104, 2020 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32664891

RESUMEN

BACKGROUND: Africa bears a disproportionately high burden of globally significant disease but has lagged in knowledge production to address its health challenges. In this contribution, we discuss the challenges and approaches to health research capacity strengthening in sub-Saharan Africa and propose that the recent shift to an African-led approach is the most optimal. METHODS AND FINDINGS: We introduce several capacity building approaches and recent achievements, explore why African-led research on the continent is a potentially paradigm-shifting and innovative approach, and discuss the advantages and challenges thereof. We reflect on the approaches used by the African Academy of Sciences (AAS)-funded Sub-Saharan African Network for TB/HIV Research Excellence (SANTHE) consortium as an example of an effective African-led science and capacity building programme. We recommend the following as crucial components of future efforts: 1. Directly empowering African-based researchers, 2. Offering quality training and career development opportunities to large numbers of junior African scientists and support staff, and 3. Effective information exchange and collaboration. Furthermore, we argue that long-term investment from international donors and increasing funding commitments from African governments and philanthropies will be needed to realise a critical mass of local capacity and to create and sustain world-class research hubs that will be conducive to address Africa's intractable health challenges. CONCLUSIONS: Our experiences so far suggest that African-led research has the potential to overcome the vicious cycle of brain-drain and may ultimately lead to improvement of health and science-led economic transformation of Africa into a prosperous continent.


Asunto(s)
Investigación Biomédica/organización & administración , Investigación Biomédica/estadística & datos numéricos , Creación de Capacidad , Intercambio de Información en Salud , Colaboración Intersectorial , Investigadores/educación , Adulto , África del Sur del Sahara , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos de Investigación
6.
Commun Biol ; 2: 203, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31231693

RESUMEN

Mucosal-associated invariant T (MAIT) cells typically express a TRAV1-2+ semi-invariant TCRα that enables recognition of bacterial, mycobacterial, and fungal riboflavin metabolites presented by MR1. MAIT cells are associated with immune control of bacterial and mycobacterial infections in murine models. Here, we report that a population of pro-inflammatory TRAV1-2+ CD8+ T cells are present in the airways and lungs of healthy individuals and are enriched in bronchoalveolar fluid of patients with active pulmonary tuberculosis (TB). High-throughput T cell receptor analysis reveals oligoclonal expansions of canonical and donor-unique TRAV1-2+ MAIT-consistent TCRα sequences within this population. Some of these cells demonstrate MR1-restricted mycobacterial reactivity and phenotypes suggestive of MAIT cell identity. These findings demonstrate enrichment of TRAV1-2+ CD8+ T cells with MAIT or MAIT-like features in the airways during active TB and suggest a role for these cells in the human pulmonary immune response to Mycobacterium tuberculosis.


Asunto(s)
Linfocitos T CD8-positivos/citología , Células T Invariantes Asociadas a Mucosa/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Bronquios/microbiología , Líquido del Lavado Bronquioalveolar , Broncoscopía , Linfocitos T CD8-positivos/microbiología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Sistema Inmunológico , Inflamación , Intestinos/inmunología , Pulmón/inmunología , Pulmón/microbiología , Ratones , Antígenos de Histocompatibilidad Menor/inmunología , Células T Invariantes Asociadas a Mucosa/microbiología , Mycobacterium tuberculosis/inmunología , Oregon , Fenotipo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Sudáfrica , Tuberculosis Pulmonar/microbiología
7.
Cytokine X ; 1(1): 100004, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33604547

RESUMEN

Host immunity is crucial for controlling M. tuberculosis infection. Functional polymorphisms in the cytokine macrophage migration inhibitory factor (MIF) show global population stratification, with the highest prevalence of low expression MIF alleles found in sub-Saharan Africans, which is a population with the greatest confluence of both TB and HIV infection and disease. We investigated the association between MIF alleles and tuberculosis (TB) and HIV in South Africa. We acquired clinical information and determined the frequency of two MIF promoter variants: a functional -794 CATT5-8 microsatellite and an associated -173 G/C SNP in two HIV-positive cohorts of patients with active laboratory-confirmed TB and in controls without active TB who were all HIV positive. We found a greater frequency of low expression MIF promoter variants (-794 CATT5,6) among TB disease cases compared to controls (OR = 2.03, p = 0.023), supporting a contribution of genetic low MIF expression to the high prevalence of TB in South Africa. Among those with HIV, circulating MIF levels also were associated with lower CD4 cell counts irrespective of TB status (p = 0.016), suggesting an influence of HIV immunosuppression on MIF expression.

8.
Tuberculosis (Edinb) ; 109: 80-84, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29559125

RESUMEN

Tuberculosis (TB) remains a serious threat in underdeveloped areas. Mycobacterium tuberculosis curli pili (MTP), a virulence factor, is a potential biomarker for a reliable point of care (POC) test and was evaluated for its ability to react with Immunoglobulin G (IgG) in TB patients. An MTP synthetic peptide in a slot blot assay was used to screen serum/plasma samples (n = 65) in 3 separate cohorts, including 40 TB positive (16 HIV co-infected), 20 TB negative/HIV negative patients and 5 healthy volunteers. Forty samples were true positives (HIV positive, n = 16), 23 true negatives (HIV negative) and 2 false positives (HIV negative). The McNemar test demonstrated a 3.08% accuracy estimate (CI: -2.1% - 3.08%). This confirms that MTP is expressed during infection, including HIV-TB co-infection, is likely to be suitable for the design of a POC test and supports the validation of MTP for TB detection in larger patient populations.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Fimbrias Bacterianas/inmunología , Inmunoensayo/métodos , Inmunoglobulina G/inmunología , Mycobacterium tuberculosis/inmunología , Fragmentos de Péptidos/inmunología , Tuberculosis Pulmonar/diagnóstico , Anticuerpos Antibacterianos/sangre , Estudios de Casos y Controles , Humanos , Inmunoglobulina G/sangre , Fragmentos de Péptidos/síntesis química , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Tuberculosis Pulmonar/sangre , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología
9.
Immunology ; 150(1): 45-54, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27633333

RESUMEN

Mucosal-associated invariant T (MAIT) cells are donor-unrestricted lymphocytes that are surprisingly abundant in humans, representing 1-10% of circulating T cells and further enriched in mucosal tissues. MAIT cells recognize and are activated by small molecule ligands produced by microbes and presented by MR1, a highly conserved MHC-related antigen-presenting protein that is ubiquitously expressed in human cells. Increasing evidence suggests that MAIT cells play a protective role in anti-bacterial immunity at mucosal interfaces. Some fungi are known to produce MAIT-activating ligands, but the role of MAIT cells in fungal infections has not yet been investigated. In viral infections, specifically HIV, which has received the most study, MAIT cell biology is clearly altered, but the mechanisms explaining these alterations and their clinical significance are not yet understood. Many questions remain unanswered about the potential of MAIT cells for protection or pathogenesis in infectious diseases. Because they interact with the universal, donor-unrestricted ligand-presenting MR1 molecule, MAIT cells may be attractive immunotherapy or vaccine targets. New tools, including the development of MR1-ligand tetramers and next-generation T-cell receptor sequencing, have the potential to accelerate MAIT cell research and lead to new insights into the role of this unique set of lymphocytes in infectious diseases.


Asunto(s)
Inmunidad Mucosa , Infecciones/inmunología , Células T Asesinas Naturales/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Diferenciación de Linfocitos B/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Antígenos de Histocompatibilidad Menor/metabolismo , Receptores de Antígenos de Linfocitos T/genética
10.
Immunity ; 44(2): 391-405, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26850658

RESUMEN

Innate lymphoid cells (ILCs) play a central role in the response to infection by secreting cytokines crucial for immune regulation, tissue homeostasis, and repair. Although dysregulation of these systems is central to pathology, the impact of HIV-1 on ILCs remains unknown. We found that human blood ILCs were severely depleted during acute viremic HIV-1 infection and that ILC numbers did not recover after resolution of peak viremia. ILC numbers were preserved by antiretroviral therapy (ART), but only if initiated during acute infection. Transcriptional profiling during the acute phase revealed upregulation of genes associated with cell death, temporally linked with a strong IFN acute-phase response and evidence of gut barrier breakdown. We found no evidence of tissue redistribution in chronic disease and remaining circulating ILCs were activated but not apoptotic. These data provide a potential mechanistic link between acute HIV-1 infection, lymphoid tissue breakdown, and persistent immune dysfunction.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/inmunología , Interferón gamma/metabolismo , Intestinos/patología , Linfocitos/inmunología , Enfermedad Aguda , Antivirales/administración & dosificación , Apoptosis/efectos de los fármacos , Apoptosis/genética , Movimiento Celular , Células Cultivadas , Enfermedad Crónica , Estudios de Cohortes , Regulación de la Expresión Génica , Infecciones por VIH/tratamiento farmacológico , Humanos , Inmunidad Innata , Interferón gamma/genética , Intestinos/virología , Linfocitos/efectos de los fármacos , Linfocitos/virología , Factores de Tiempo , Resultado del Tratamiento , Carga Viral/efectos de los fármacos , Carga Viral/inmunología
11.
J Infect Dis ; 213(4): 628-33, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26374910

RESUMEN

Single-cell analysis captures the heterogeneity of T-cell populations that target defined antigens. Human immunodeficiency virus (HIV) infection results in defects of antimycobacterial immunity, which remain poorly defined. We therefore recruited a small number of subjects, including those with latent and active M. tuberculosis infection, with or without concomitant HIV infection, and tracked the mycobacterial glycolipid-reactive T-cell repertoire by using CD1b tetramers. Glycolipid-reactive T cells expressed memory markers and the HIV coreceptors CD4 and CCR5; they were not detected in subjects with HIV-associated active M. tuberculosis infection. HIV infection may affect T cells that recognize mycobacterial glycolipids and influence immunity.


Asunto(s)
Glucolípidos/inmunología , Infecciones por VIH/inmunología , Mycobacterium/inmunología , Linfocitos T/inmunología , Tuberculosis/inmunología , Adulto , Antígenos CD4/análisis , Coinfección/inmunología , Humanos , Memoria Inmunológica , Persona de Mediana Edad , Receptores CCR5/análisis , Subgrupos de Linfocitos T/inmunología , Linfocitos T/química
12.
EBioMedicine ; 2(9): 1160-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26501113

RESUMEN

Biomarkers for active tuberculosis (TB) are urgently needed to improve rapid TB diagnosis. The objective of this study was to identify serum protein expression changes associated with TB but not latent Mycobacterium tuberculosis infection (LTBI), uninfected states, or respiratory diseases other than TB (ORD). Serum samples from 209 HIV uninfected (HIV(-)) and co-infected (HIV(+)) individuals were studied. In the discovery phase samples were analyzed via liquid chromatography and mass spectrometry, and in the verification phase biologically independent samples were analyzed via a multiplex multiple reaction monitoring mass spectrometry (MRM-MS) assay. Compared to LTBI and ORD, host proteins were significantly differentially expressed in TB, and involved in the immune response, tissue repair, and lipid metabolism. Biomarker panels whose composition differed according to HIV status, and consisted of 8 host proteins in HIV(-) individuals (CD14, SEPP1, SELL, TNXB, LUM, PEPD, QSOX1, COMP, APOC1), or 10 host proteins in HIV(+) individuals (CD14, SEPP1, PGLYRP2, PFN1, VASN, CPN2, TAGLN2, IGFBP6), respectively, distinguished TB from ORD with excellent accuracy (AUC = 0.96 for HIV(-) TB, 0.95 for HIV(+) TB). These results warrant validation in larger studies but provide promise that host protein biomarkers could be the basis for a rapid, blood-based test for TB.


Asunto(s)
Biomarcadores/sangre , Coinfección/complicaciones , Infecciones por VIH/complicaciones , Tuberculosis/complicaciones , Adulto , Área Bajo la Curva , Proteínas Sanguíneas/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tuberculosis/sangre
13.
EBioMedicine ; 2(4): 334-340, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26114158

RESUMEN

In recent years, chronic immune activation and systemic inflammation have emerged as hallmarks of HIV disease progression and mortality. Several studies indicate that soluble inflammatory biomarkers (sCD14, IL-6, IL-8, CRP and hyaluronic acid), as well as surface markers of T-cell activation (CD38, HLA-DR) independently predict progression to AIDS and mortality in HIV-infected individuals. While co-infections have been shown to contribute to immune activation, the impact of latent tuberculosis infection (LTBI), which is widely endemic in the areas most affected by the global AIDS epidemic, has not been evaluated. We hypothesized that both active and latent states of Mycobacterium tuberculosis co-infection contribute to elevated immune activation as measurable by these markers. In HIV-infected individuals with active, but not latent TB, we found elevated levels of soluble markers associated with monocyte activation. Interestingly, T-cell activation was elevated individuals with both latent and active TB. These results suggest that in the highly TB- and HIV-endemic settings of southern Africa, latent TB-associated T-cell activation may contribute to HIV disease progression and exacerbate the HIV epidemic. In addition, our findings indicate that aggressive campaigns to treat LTBI in HIV-infected individuals in high-burden countries will not only impact TB rates, but may also slow HIV progression.

14.
PLoS One ; 10(3): e0118654, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781898

RESUMEN

The ability of antigen-specific T cells to simultaneously produce multiple cytokines is thought to correlate with the functional capacity and efficacy of T cells. These 'polyfunctional' T cells have been associated with control of HIV. We aimed to assess the impact of co-infection with Mycobacterium tuberculosis (MTB) on HIV-specific CD8+ and CD4+ T cell function. We assessed T cell functionality in 34 South African adults by investigating the IFN-y, IL-2, TNF-α, IL-21 and IL-17 cytokine secretion capacity, using polychromatic flow cytometry, following HIV Gag-specific stimulation of peripheral blood mononuclear cells. We show that MTB is associated with lower HIV-specific T cell function in co-infected as compared to HIV mono-infected individuals. This decline in function was greatest in co-infection with active Tuberculosis (TB) compared to co-infection with latent MTB (LTBI), suggesting that mycobacterial load may contribute to this loss of function. The described impact of MTB on HIV-specific T cell function may be a mechanism for increased HIV disease progression in co-infected subjects as functionally impaired T cells may be less able to control HIV.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , VIH/inmunología , Mycobacterium tuberculosis/inmunología , Adulto , Coinfección/inmunología , Femenino , Citometría de Flujo , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Humanos , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucinas/metabolismo , Tuberculosis Latente/complicaciones , Tuberculosis Latente/inmunología , Leucocitos Mononucleares/inmunología , Masculino , Tuberculosis/complicaciones , Tuberculosis/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
15.
Immunology ; 145(3): 443-53, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25752900

RESUMEN

Mucosa-associated invariant T (MAIT) cells express the semi-invariant T-cell receptor TRAV1-2 and detect a range of bacteria and fungi through the MHC-like molecule MR1. However, knowledge of the function and phenotype of bacteria-reactive MR1-restricted TRAV1-2(+) MAIT cells from human blood is limited. We broadly characterized the function of MR1-restricted MAIT cells in response to bacteria-infected targets and defined a phenotypic panel to identify these cells in the circulation. We demonstrated that bacteria-reactive MR1-restricted T cells shared effector functions of cytolytic effector CD8(+) T cells. By analysing an extensive panel of phenotypic markers, we determined that CD26 and CD161 were most strongly associated with these T cells. Using FACS to sort phenotypically defined CD8(+) subsets we demonstrated that high expression of CD26 on CD8(+)  TRAV1-2(+) cells identified with high specificity and sensitivity, bacteria-reactive MR1-restricted T cells from human blood. CD161(hi) was also specific for but lacked sensitivity in identifying all bacteria-reactive MR1-restricted T cells, some of which were CD161(dim) . Using cell surface expression of CD8, TRAV1-2, and CD26(hi) in the absence of stimulation we confirm that bacteria-reactive T cells are lacking in the blood of individuals with active tuberculosis and are restored in the blood of individuals undergoing treatment for tuberculosis.


Asunto(s)
Dipeptidil Peptidasa 4/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Membrana Mucosa/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Células Cultivadas , Dipeptidil Peptidasa 4/metabolismo , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Antígenos de Histocompatibilidad Menor , Mycobacterium smegmatis/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
16.
AIDS ; 28(18): 2671-6, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25211438

RESUMEN

OBJECTIVES: This study aimed to assess how Mycobacterium tuberculosis (MTB) coinfection alters the impact of interleukin-10 in chronic HIV infection. DESIGN: We assessed plasma cytokine levels (interleukin-10, interferon-γ, tumor necrosis factor-α, interleukin-2, interleukin-6 and interleukin-13) in 82 individuals presenting with HIV monoinfection, HIV-LTBI (latent MTB infection) coinfection or HIV-TB (active tuberculosis) coinfection. We also assessed the influence of MTB on the functional impact of interleukin-10 receptor alpha (interleukin-10Rα) blockade on HIV and MTB-specific CD4(+) T cells. METHODS: Plasma cytokine levels were measured by high sensitivity Luminex. We used an ex-vivo interleukin-10Rα blockade assay to assess if functional enhancement of HIV and MTB-specific CD4(+) T cells was possible following a 48-h stimulation with HIV gag or pooled ESAT-6 (6 kDa early secretory antigenic target) and CFP-10 (10-kDa culture filtrate protein) peptides. Cell supernatant was collected 48 h after stimulation and the cytokine profile was measured by Luminex. RESULTS: Plasma interleukin-10 levels were elevated in HIV-TB as compared with HIV monoinfection (P < 0.05) and HIV-LTBI (P < 0.05). Plasma interleukin-10 levels correlated to HIV viral load in HIV monoinfection (P = 0.016) and HIV-LTBI (P = 0.042), but not HIV-TB. Ex-vivo blockade of interleukin-10Rα significantly enhanced MTB and HIV-specific CD4(+) T-cell function in HIV-LTBI individuals but not in HIV-TB individuals. CONCLUSION: Tuberculosis disrupts the correlation between interleukin-10 and markers of HIV disease progression. In addition, HIV-TB is associated with a more inflammatory cytokine milieu compared with HIV monoinfection. Interestingly, interleukin-10Rα blockade can enhance both HIV and MTB-specific T-cell function in HIV-LTBI, but not in HIV-TB coinfection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Interleucina-10/sangre , Mycobacterium tuberculosis/inmunología , Tuberculosis/complicaciones , Tuberculosis/inmunología , Humanos , Interleucina-10/inmunología
17.
Clin Vaccine Immunol ; 21(6): 791-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24671553

RESUMEN

Serology data are limited for patients with sputum smear-negative HIV-associated active tuberculosis (TB). We evaluated the serum antibody responses against the mycobacterial proteins MPT51, MS, and echA1 and the 38-kDa protein via enzyme-linked immunosorbent assay (ELISA) in South African (S.A.) HIV-positive (HIV(+)) smear-negative TB patients (n = 56), U.S. HIV(+) controls with a positive tuberculin skin test (TST(+); n = 21), and S.A. HIV-negative (HIV(-)) (n = 18) and HIV(+) (n = 24) controls. TB patients had positive antibody reactivity against MPT51 (73%), echA1 (59%), MS (36%), and the 38-kDa protein (11%). Little reactivity against MPT51 and echA1 was observed in control groups at low risk for TB, i.e., S.A. HIV(-) (0% and 6%, respectively), and at moderate risk for TB development, i.e., U.S. HIV(+) TST(+) controls (14% and 10%, respectively). By contrast, more reactivity was detected in the S.A. HIV(+) control group at higher risk for TB (25% and 45%, respectively). Our data hold promise that antibody detection against MPT51 and echA1 might have adjunctive value in the detection of HIV(+) smear-negative TB and might reflect increasing Mycobacterium tuberculosis infection activity in asymptomatic HIV(+) individuals.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Lipoproteínas/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/diagnóstico , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Femenino , Infecciones por VIH/sangre , Infecciones por VIH/inmunología , Humanos , Malato Sintasa/inmunología , Masculino , Persona de Mediana Edad , Sudáfrica , Esputo/microbiología , Esputo/virología , Prueba de Tuberculina , Tuberculosis Pulmonar/inmunología
18.
J Virol ; 88(8): 4291-303, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478420

RESUMEN

UNLABELLED: The antiviral role of TRIM E3 ligases in vivo is not fully understood. To test the hypothesis that TRIM5α and TRIM22 have differential transcriptional regulation and distinct anti-HIV roles according to infection phase and compartment, we measured TRIM5α, TRIM22, and type I interferon (IFN-I)-inducible myxovirus resistance protein A (MxA) levels in peripheral blood mononuclear cells (PBMCs) during primary and chronic HIV-1 infection, with chronic infection samples being matched PBMCs and central nervous system (CNS)-derived cells. Associations with biomarkers of disease progression were explored. The impact of IFN-I, select proinflammatory cytokines, and HIV on TRIM E3 ligase-specific expression was investigated. PBMCs from individuals with primary and chronic HIV-1 infection had significantly higher levels of MxA and TRIM22 than did PBMCs from HIV-1-negative individuals (P < 0.05 for all comparisons). PBMCs from chronic infection had lower levels of TRIM5α than did PBMCs from primary infection or HIV-1-uninfected PBMCs (P = 0.0001 for both). In matched CNS-derived samples and PBMCs, higher levels of MxA (P = 0.001) and TRIM5α (P = 0.0001) in the CNS were noted. There was a negative correlation between TRIM22 levels in PBMCs and plasma viral load (r = -0.40; P = 0.04). In vitro, IFN-I and, rarely, proinflammatory cytokines induced TRIM5α and TRIM22 in a cell type-dependent manner, and the knockdown of either protein in CD4(+) lymphocytes resulted in increased HIV-1 infection. These data suggest that there are infection-phase-specific and anatomically compartmentalized differences in TRIM5α and TRIM22 regulation involving primarily IFN-I and specific cell types and indicate subtle differences in the antiviral roles and transcriptional regulation of TRIM E3 ligases in vivo. IMPORTANCE: Type I interferon-inducible TRIM E3 ligases are a family of intracellular proteins with potent antiviral activities mediated through diverse mechanisms. However, little is known about the contribution of these proteins to antiviral immunity in vivo and how their expression is regulated. We show here that TRIM5α and TRIM22, two prominent members of the family, have different expression patterns in vivo and that the expression pattern depends on HIV-1 infection status and phase. Furthermore, expression differs in peripheral blood versus central nervous system anatomical sites of infection. Only TRIM22 expression correlated negatively with HIV-1 viral load, but gene silencing of both proteins enhances HIV-1 infection of target cells. We report subtle differences in TRIM5α and TRIM22 gene induction by IFN-I and proinflammatory cytokines in CD4(+) lymphocytes, monocytes, and neuronal cells. This study enhances our understanding of antiviral immunity by intrinsic antiviral factors and how their expression is determined.


Asunto(s)
Proteínas Portadoras/metabolismo , Infecciones por VIH/metabolismo , VIH-1/fisiología , Proteínas Represoras/metabolismo , Adolescente , Adulto , Factores de Restricción Antivirales , Proteínas Portadoras/genética , Niño , Estudios de Cohortes , Femenino , Regulación de la Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Proteínas Represoras/genética , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Replicación Viral , Adulto Joven
19.
Am J Respir Crit Care Med ; 188(9): 1137-46, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24047412

RESUMEN

RATIONALE: A hallmark of pulmonary tuberculosis (TB) is the formation of granulomas. However, the immune factors that drive the formation of a protective granuloma during latent TB, and the factors that drive the formation of inflammatory granulomas during active TB, are not well defined. OBJECTIVES: The objective of this study was to identify the underlying immune mechanisms involved in formation of inflammatory granulomas seen during active TB. METHODS: The immune mediators involved in inflammatory granuloma formation during TB were assessed using human samples and experimental models of Mycobacterium tuberculosis infection, using molecular and immunologic techniques. MEASUREMENTS AND MAIN RESULTS: We demonstrate that in human patients with active TB and in nonhuman primate models of M. tuberculosis infection, neutrophils producing S100 proteins are dominant within the inflammatory lung granulomas seen during active TB. Using the mouse model of TB, we demonstrate that the exacerbated lung inflammation seen as a result of neutrophilic accumulation is dependent on S100A8/A9 proteins. S100A8/A9 proteins promote neutrophil accumulation by inducing production of proinflammatory chemokines and cytokines, and influencing leukocyte trafficking. Importantly, serum levels of S100A8/A9 proteins along with neutrophil-associated chemokines, such as keratinocyte chemoattractant, can be used as potential surrogate biomarkers to assess lung inflammation and disease severity in human TB. CONCLUSIONS: Our results thus show a major pathologic role for S100A8/A9 proteins in mediating neutrophil accumulation and inflammation associated with TB. Thus, targeting specific molecules, such as S100A8/A9 proteins, has the potential to decrease lung tissue damage without impacting protective immunity against TB.


Asunto(s)
Calgranulina A/inmunología , Calgranulina B/inmunología , Granuloma del Sistema Respiratorio/inmunología , Mediadores de Inflamación/inmunología , Neutrófilos/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Quimiocinas/inmunología , Factores Quimiotácticos/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL
20.
J Infect Dis ; 208(10): 1571-9, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23922364

RESUMEN

BACKGROUND: Tuberculosis is transmitted by patients with pulmonary disease. Matrix metalloproteinases (MMPs) drive lung destruction in tuberculosis but the resulting matrix degradation products (MDPs) have not been studied. We investigate the hypothesis that MMP activity generates matrix turnover products as correlates of lung pathology. METHODS: Induced sputum and plasma were collected prospectively from human immunodeficiency virus (HIV) positive and negative patients with pulmonary tuberculosis and controls. Concentrations of MDPs and MMPs were analyzed by ELISA and Luminex array in 2 patient cohorts. RESULTS: Procollagen III N-terminal propeptide (PIIINP) was 3.8-fold higher in induced sputum of HIV-uninfected tuberculosis patients compared to controls and desmosine, released during elastin degradation, was 2.4-fold higher. PIIINP was elevated in plasma of tuberculosis patients. Plasma PIIINP correlated with induced sputum MMP-1 concentrations and radiological scores, demonstrating that circulating MDPs reflect lung destruction. In a second patient cohort of mixed HIV seroprevalence, plasma PIIINP concentration was increased 3.0-fold above controls (P < .001). Plasma matrix metalloproteinase-8 concentrations were also higher in tuberculosis patients (P = .001). Receiver operating characteristic analysis utilizing these 2 variables demonstrated an area under the curve of 0.832 (P < .001). CONCLUSIONS: In pulmonary tuberculosis, MMP-driven immunopathology generates matrix degradation products.


Asunto(s)
Desmosina/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Fragmentos de Péptidos/metabolismo , Procolágeno/metabolismo , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/patología , Biomarcadores , Coinfección , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Seropositividad para VIH , Metaloproteinasas de la Matriz/sangre , Fragmentos de Péptidos/sangre , Procolágeno/sangre , Curva ROC , Reproducibilidad de los Resultados , Esputo/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...