Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Curr Opin HIV AIDS ; 19(4): 201-211, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38841850

ABSTRACT

PURPOSE OF REVIEW: Highlighting opportunities/potential for immunotherapy by understanding dynamics of HIV control during pediatric HIV infection with and without antiretroviral therapy (ART), as modeled in Simian immunodeficiency virus (SIV) and Simian-human immunodeficiency virus (SHIV)-infected rhesus macaques and observed in clinical trials. This review outlines mode of transmission, pathogenesis of pediatric HIV, unique aspects of the infant immune system, infant macaque models and immunotherapies. RECENT FINDINGS: During the earliest stages of perinatal HIV infection, the infant immune system is characterized by a unique environment defined by immune tolerance and lack of HIV-specific T cell responses which contribute to disease progression. Moreover, primary lymphoid organs such as the thymus appear to play a distinct role in HIV pathogenesis in children living with HIV (CLWH). Key components of the immune system determine the degree of viral control, targets for strategies to induce viral control, and the response to immunotherapy. The pursuit of highly potent broadly neutralizing antibodies (bNAbs) and T cell vaccines has revolutionized the approach to HIV cure. Administration of HIV-1-specific bNAbs, targeting the highly variable envelope improves humoral immunity, and T cell vaccines induce or improve T cell responses such as the cytotoxic effects of HIV-1-specific CD8+ T cells, both of which are promising options towards virologic control and ART-free remission as evidenced by completed and ongoing clinical trials. SUMMARY: Understanding early events during HIV infection and disease progression in CLWH serves as a foundation for predicting or targeting later outcomes by harnessing the immune system's natural responses. The developing pediatric immune system offers multiple opportunities for specific long-term immunotherapies capable of improving quality of life during adolescence and adulthood.


Subject(s)
HIV Infections , Immunotherapy , Humans , HIV Infections/immunology , Immunotherapy/methods , Animals , Child , Macaca mulatta , Disease Models, Animal , Infant , Simian Immunodeficiency Virus/immunology , AIDS Vaccines/immunology , AIDS Vaccines/administration & dosage
2.
bioRxiv ; 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38293221

ABSTRACT

Although vaccines have reduced COVID-19 disease burden, their efficacy in helminth infection endemic areas is not well characterized. We evaluated the impact of infection by Heligmosomoides polygyrus bakeri (Hpb), a murine intestinal hookworm, on the efficacy of an mRNA vaccine targeting the Wuhan-1 spike protein of SARS-CoV-2. Although immunization generated similar B cell responses in Hpb-infected and uninfected mice, polyfunctional CD4+ and CD8+ T cell responses were markedly reduced in Hpb-infected mice. Hpb-infected and mRNA vaccinated mice were protected against the ancestral SARS-CoV-2 strain WA1/2020, but control of lung infection was diminished against an Omicron variant compared to animals immunized without Hpb infection. Helminth mediated suppression of spike-specific CD8+ T cell responses occurred independently of STAT6 signaling, whereas blockade of IL-10 rescued vaccine-induced CD8+ T cell responses. In mice, intestinal helminth infection impairs vaccine induced T cell responses via an IL-10 pathway and compromises protection against antigenically shifted SARS-CoV-2 variants.

3.
Nat Commun ; 14(1): 1914, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024448

ABSTRACT

The immunopathological mechanisms driving the development of severe COVID-19 remain poorly defined. Here, we utilize a rhesus macaque model of acute SARS-CoV-2 infection to delineate perturbations in the innate immune system. SARS-CoV-2 initiates a rapid infiltration of plasmacytoid dendritic cells into the lower airway, commensurate with IFNA production, natural killer cell activation, and a significant increase of blood CD14-CD16+ monocytes. To dissect the contribution of lung myeloid subsets to airway inflammation, we generate a longitudinal scRNA-Seq dataset of airway cells, and map these subsets to corresponding populations in the human lung. SARS-CoV-2 infection elicits a rapid recruitment of two macrophage subsets: CD163+MRC1-, and TREM2+ populations that are the predominant source of inflammatory cytokines. Treatment with baricitinib (Olumiant®), a JAK1/2 inhibitor is effective in eliminating the influx of non-alveolar macrophages, with a reduction of inflammatory cytokines. This study delineates the major lung macrophage subsets driving airway inflammation during SARS-CoV-2 infection.


Subject(s)
COVID-19 , Animals , Humans , Macaca mulatta , SARS-CoV-2 , Macrophages , Inflammation , Cytokines , Membrane Glycoproteins , Receptors, Immunologic
4.
5.
J Clin Invest ; 132(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-35230978

ABSTRACT

Interleukin-10 (IL-10) is an immunosuppressive cytokine that signals through STAT3 to regulate T follicular helper (Tfh) cell differentiation and germinal center formation. In SIV-infected macaques, levels of IL-10 in plasma and lymph nodes (LNs) were induced by infection and not normalized with antiretroviral therapy (ART). During chronic infection, plasma IL-10 and transcriptomic signatures of IL-10 signaling were correlated with the cell-associated SIV-DNA content within LN CD4+ memory subsets, including Tfh cells, and predicted the frequency of CD4+ Tfh cells and their cell-associated SIV-DNA content during ART, respectively. In ART-treated rhesus macaques, cells harboring SIV-DNA by DNAscope were preferentially found in the LN B cell follicle in proximity to IL-10. Finally, we demonstrated that the in vivo neutralization of soluble IL-10 in ART-treated, SIV-infected macaques reduced B cell follicle maintenance and, by extension, LN memory CD4+ T cells, including Tfh cells and those expressing PD-1 and CTLA-4. Thus, these data support a role for IL-10 in maintaining a pool of target cells in lymphoid tissue that serve as a niche for viral persistence. Targeting IL-10 signaling to impair CD4+ T cell survival and improve antiviral immune responses may represent a novel approach to limit viral persistence in ART-suppressed people living with HIV.


Subject(s)
HIV Infections , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , Humans , Interleukin-10/genetics , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy
6.
bioRxiv ; 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34642693

ABSTRACT

The COVID-19 pandemic remains a global health crisis, yet, the immunopathological mechanisms driving the development of severe disease remain poorly defined. Here, we utilize a rhesus macaque (RM) model of SARS-CoV-2 infection to delineate perturbations in the innate immune system during acute infection using an integrated systems analysis. We found that SARS-CoV-2 initiated a rapid infiltration (two days post infection) of plasmacytoid dendritic cells into the lower airway, commensurate with IFNA production, natural killer cell activation, and induction of interferon-stimulated genes. At this early interval, we also observed a significant increase of blood CD14-CD16+ monocytes. To dissect the contribution of lung myeloid subsets to airway inflammation, we generated a novel compendium of RM-specific lung macrophage gene expression using a combination of sc-RNA-Seq data and bulk RNA-Seq of purified populations under steady state conditions. Using these tools, we generated a longitudinal sc-RNA-seq dataset of airway cells in SARS-CoV-2-infected RMs. We identified that SARS-CoV-2 infection elicited a rapid recruitment of two subsets of macrophages into the airway: a C206+MRC1-population resembling murine interstitial macrophages, and a TREM2+ population consistent with CCR2+ infiltrating monocytes, into the alveolar space. These subsets were the predominant source of inflammatory cytokines, accounting for ~75% of IL6 and TNF production, and >90% of IL10 production, whereas the contribution of CD206+MRC+ alveolar macrophages was significantly lower. Treatment of SARS-CoV-2 infected RMs with baricitinib (Olumiant ® ), a novel JAK1/2 inhibitor that recently received Emergency Use Authorization for the treatment of hospitalized COVID-19 patients, was remarkably effective in eliminating the influx of infiltrating, non-alveolar macrophages in the alveolar space, with a concomitant reduction of inflammatory cytokines. This study has delineated the major subsets of lung macrophages driving inflammatory and anti-inflammatory cytokine production within the alveolar space during SARS-CoV-2 infection. ONE SENTENCE SUMMARY: Multi-omic analyses of hyperacute SARS-CoV-2 infection in rhesus macaques identified two population of infiltrating macrophages, as the primary orchestrators of inflammation in the lower airway that can be successfully treated with baricitinib.

7.
Cell ; 184(2): 460-475.e21, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33278358

ABSTRACT

SARS-CoV-2-induced hypercytokinemia and inflammation are critically associated with COVID-19 severity. Baricitinib, a clinically approved JAK1/JAK2 inhibitor, is currently being investigated in COVID-19 clinical trials. Here, we investigated the immunologic and virologic efficacy of baricitinib in a rhesus macaque model of SARS-CoV-2 infection. Viral shedding measured from nasal and throat swabs, bronchoalveolar lavages, and tissues was not reduced with baricitinib. Type I interferon (IFN) antiviral responses and SARS-CoV-2-specific T cell responses remained similar between the two groups. Animals treated with baricitinib showed reduced inflammation, decreased lung infiltration of inflammatory cells, reduced NETosis activity, and more limited lung pathology. Importantly, baricitinib-treated animals had a rapid and remarkably potent suppression of lung macrophage production of cytokines and chemokines responsible for inflammation and neutrophil recruitment. These data support a beneficial role for, and elucidate the immunological mechanisms underlying, the use of baricitinib as a frontline treatment for inflammation induced by SARS-CoV-2 infection.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Azetidines/administration & dosage , COVID-19 Drug Treatment , COVID-19/immunology , Macaca mulatta , Neutrophil Infiltration/drug effects , Purines/administration & dosage , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Animals , COVID-19/physiopathology , Cell Death/drug effects , Cell Degranulation/drug effects , Disease Models, Animal , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Janus Kinases/antagonists & inhibitors , Lung/drug effects , Lung/immunology , Lung/pathology , Lymphocyte Activation/drug effects , Macrophages, Alveolar/immunology , SARS-CoV-2/physiology , Severity of Illness Index , T-Lymphocytes/immunology , Virus Replication/drug effects
8.
bioRxiv ; 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32995780

ABSTRACT

Effective therapeutics aimed at mitigating COVID-19 symptoms are urgently needed. SARS-CoV-2 induced hypercytokinemia and systemic inflammation are associated with disease severity. Baricitinib, a clinically approved JAK1/2 inhibitor with potent anti-inflammatory properties is currently being investigated in COVID-19 human clinical trials. Recent reports suggest that baricitinib may also have antiviral activity in limiting viral endocytosis. Here, we investigated the immunologic and virologic efficacy of baricitinib in a rhesus macaque model of SARS-CoV-2 infection. Viral shedding measured from nasal and throat swabs, bronchoalveolar lavages and tissues was not reduced with baricitinib. Type I IFN antiviral responses and SARS-CoV-2 specific T cell responses remained similar between the two groups. Importantly, however, animals treated with baricitinib showed reduced immune activation, decreased infiltration of neutrophils into the lung, reduced NETosis activity, and more limited lung pathology. Moreover, baricitinib treated animals had a rapid and remarkably potent suppression of alveolar macrophage derived production of cytokines and chemokines responsible for inflammation and neutrophil recruitment. These data support a beneficial role for, and elucidate the immunological mechanisms underlying, the use of baricitinib as a frontline treatment for severe inflammation induced by SARS-CoV-2 infection.

9.
Immunity ; 47(4): 776-788.e5, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045906

ABSTRACT

Antiretroviral therapy (ART) suppresses viral replication in HIV-infected individuals but does not eliminate the reservoir of latently infected cells. Recent work identified PD-1+ follicular helper T (Tfh) cells as an important cellular compartment for viral persistence. Here, using ART-treated, SIV-infected rhesus macaques, we show that CTLA-4+PD-1- memory CD4+ T cells, which share phenotypic markers with regulatory T cells, were enriched in SIV DNA in blood, lymph nodes (LN), spleen, and gut, and contained replication-competent and infectious virus. In contrast to PD-1+ Tfh cells, SIV-enriched CTLA-4+PD-1- CD4+ T cells were found outside the B cell follicle of the LN, predicted the size of the persistent viral reservoir during ART, and significantly increased their contribution to the SIV reservoir with prolonged ART-mediated viral suppression. We have shown that CTLA-4+PD-1- memory CD4+ T cells are a previously unrecognized component of the SIV and HIV reservoir that should be therapeutically targeted for a functional HIV-1 cure.


Subject(s)
Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CTLA-4 Antigen/immunology , Programmed Cell Death 1 Receptor/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CTLA-4 Antigen/metabolism , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immunologic Memory/drug effects , Immunologic Memory/immunology , In Situ Hybridization , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/virology , Macaca mulatta , Microscopy, Confocal , Programmed Cell Death 1 Receptor/metabolism , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/virology
10.
Parasitol Int ; 65(6 Pt A): 702-707, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27546158

ABSTRACT

The production of ergosterol lipid, important for the Leishmania membrane homeostasis, involves different enzymes. This pathway can be blocked to azoles and allylamines drugs, such as Butenafine. The aim of the present work was to evaluate the anti-leishmanicidal activity of this drug in 2 major species of Leishmania responsible for causing the American tegumentar leishmaniasis (L. (L.) amazonensis and L. (V.) braziliensis). Butenafine eliminated promastigote forms of L. amazonensis and L. braziliensis with efficacy similar to miltefosine, a standard anti-leishmania drug. In addition, butenafine induced alterations in promastigote forms of L. amazonensis that resemble programmed cell death. Butenafine as well as miltefosine presented mild toxicity in peritoneal macrophages, however, butenafine was more effective to eliminate intracellular amastigotes of both L. amazonensis and L. braziliensis, and this effect was not associated with elevated levels of nitric oxide or hydrogen peroxide. Taken together, data presented herein suggests that butenafine can be considered as a prototype drug able to eliminate L. amazonensis and L. braziliensis, etiological agents of anergic diffuse and mucocutaneous leishmaniasis, respectively.


Subject(s)
Antifungal Agents/therapeutic use , Antiprotozoal Agents/therapeutic use , Benzylamines/therapeutic use , Leishmania braziliensis/drug effects , Leishmaniasis, Diffuse Cutaneous/drug therapy , Leishmaniasis, Mucocutaneous/drug therapy , Naphthalenes/therapeutic use , Phosphorylcholine/analogs & derivatives , Animals , Drug Repositioning , Female , Leishmania braziliensis/classification , Leishmaniasis, Diffuse Cutaneous/parasitology , Leishmaniasis, Mucocutaneous/parasitology , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Parasitic Sensitivity Tests , Phosphorylcholine/therapeutic use
11.
PLoS One ; 10(12): e0144946, 2015.
Article in English | MEDLINE | ID: mdl-26674781

ABSTRACT

Among neglected tropical diseases, leishmaniasis is one of the most important ones, affecting more than 12 million people worldwide. The available treatments are not well tolerated, and present diverse side effects, justifying the search for new therapeutic compounds. In the present study, the activity of ursolic acid (UA) and oleanolic acid (OA) were assayed in experimental cutaneous leishmaniasis (in vitro and in vivo). Promastigote forms of L. amazonensis were incubated with OA and UA for 24h, and effective concentration 50% (EC50) was estimated. Ultraestructural alterations in Leishmania amazonensis promastigotes after UA treatment were evaluated by transmission electron microscopy, and the possible mode of action was assayed through Annexin V and propidium iodide staining, caspase 3/7 activity, DNA fragmentation and transmembrane mitochondrial potential. The UA potential was evaluated in intracellular amastigotes, and its therapeutic potential was evaluated in L. amazonensis infected BALB/c mice. UA eliminated L. amazonensis promastigotes with an EC50 of 6.4 µg/mL, comparable with miltefosine, while OA presented only a marginal effect on promastigote forms at 100 µg/mL. The possible mechanism by which promastigotes were eliminated by UA was programmed cell death, independent of caspase 3/7, but it was highly dependent on mitochondria activity. UA was not toxic for peritoneal macrophages from BALB/c mice, and it was able to eliminate intracellular amastigotes, associated with nitric oxide (NO) production. OA did not eliminate amastigotes nor trigger NO. L. amazonensis infected BALB/c mice submitted to UA treatment presented lesser lesion size and parasitism compared to control. This study showed, for the first time, that UA eliminate promastigote forms through a mechanism associated with programed cell death, and importantly, was effective in vivo. Therefore, UA can be considered an interesting candidate for future tests as a prototype drug for the treatment of cutaneous leishmaniasis.


Subject(s)
Apoptosis/drug effects , Leishmania/drug effects , Leishmaniasis, Cutaneous/parasitology , Triterpenes/pharmacology , Trypanocidal Agents/pharmacology , Animals , Cytokines/biosynthesis , Disease Models, Animal , Female , Leishmania/ultrastructure , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Cutaneous/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/parasitology , Male , Mice , Nitric Oxide/metabolism , Oleanolic Acid/pharmacology , Plant Extracts/pharmacology , Ursolic Acid
12.
PLoS One ; 9(10): e108984, 2014.
Article in English | MEDLINE | ID: mdl-25303100

ABSTRACT

BACKGROUND: The participation of immune/inflammatory mechanisms in the pathogenesis of tropical endomyocardial fibrosis (EMF) has been suggested by the finding of early blood and myocardial eosinophilia. However, the inflammatory activation status of late-stage EMF patients is still unknown. METHODOLOGY/PRINCIPAL FINDINGS: We evaluated pro- and anti-inflammatory cytokine levels in plasma samples from late stage EMF patients. Cytokine levels of Tumor Necrosis Factor (TNF)-α, Interferon (IFN)-γ, Interleukin (IL)-2, IL-4, IL-6, and IL-10 were assayed in plasma samples from 27 EMF patients and compared with those of healthy control subjects. All EMF patients displayed detectable plasma levels of at least one of the cytokines tested. We found that TNF-α, IL-6, IL-4, and IL-10 were each detected in at least 74% of tested sera, and plasma levels of IL-10, IL-4, and TNF-α were significantly higher than those of controls. Plasma levels of such cytokines positively correlated with each other. CONCLUSIONS/SIGNIFICANCE: The mixed pro- and anti-inflammatory/Th2circulating cytokine profile in EMF is consistent with the presence of a persistent inflammatory stimulus. On the other hand, the detection of increased levels of TNF-α may be secondary to the cardiovascular involvement observed in these patients, whereas IL-4 and IL-10 may have been upregulated as a homeostatic mechanism to buffer both production and deleterious cardiovascular effects of pro-inflammatory cytokines. Further studies might establish whether these findings play a role in disease pathogenesis.


Subject(s)
Endomyocardial Fibrosis/blood , Interleukin-10/blood , Interleukin-4/blood , Tumor Necrosis Factor-alpha/blood , Adult , Female , Humans , Interferon-gamma/blood , Interleukin-2/blood , Interleukin-6/blood , Male , Middle Aged , Young Adult
13.
J Virol ; 88(23): 13836-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253339

ABSTRACT

UNLABELLED: Memory stem T cells (T(SCM)) constitute a long-lived, self-renewing lymphocyte population essential for the maintenance of functional immunity. The hallmarks of HIV-1 pathogenesis are CD4(+) T cell depletion and abnormal cellular activation. We investigated the impact of HIV-1 infection on the T(SCM) compartment, as well as any protective role these cells may have in disease progression, by characterizing this subset in a cohort of 113 subjects with various degrees of viral control on and off highly active antiretroviral therapy (HAART). We observed that the frequency of CD8(+) T(SCM) was decreased in all individuals with chronic, untreated HIV-1 infection and that HAART had a restorative effect on this subset. In contrast, natural controllers of HIV-1 had the highest absolute number of CD4(+) T(SCM) cells among all of the infected groups. The frequency of CD4(+) T(SCM) predicted higher CD8(+) T(SCM) frequencies, consistent with a role for the CD4(+) subset in helping to maintain CD8(+) memory T cells. In addition, T(SCM) appeared to be progenitors for effector T cells (TEM), as these two compartments were inversely correlated. Increased frequencies of CD8(+) T(SCM) predicted lower viral loads, higher CD4(+) counts, and less CD8(+) T cell activation. Finally, we found that T(SCM) express the mucosal homing integrin α4ß7 and can be identified in gut-associated lymphoid tissue (GALT). The frequency of mucosal CD4(+) T(SCM) was inversely correlated with that in the blood, potentially reflecting the ability of these self-renewing cells to migrate to a crucial site of ongoing viral replication and CD4(+) T cell depletion. IMPORTANCE: HIV-1 infection leads to profound impairment of the immune system. T(SCM) constitute a recently identified lymphocyte subset with stem cell-like qualities, including the ability to generate other memory T cell subtypes, and are therefore likely to play an important role in controlling viral infection. We investigated the relationship between the size of the CD8(+) T(SCM) compartment and HIV-1 disease progression in a cohort of chronically infected individuals. Our results suggest that HAART restores a normal frequency of CD8(+) T(SCM) and that the natural preservation of this subset in the setting of untreated HIV-1 infection is associated with improved viral control and immunity. Therefore, the CD8(+) T(SCM) population may represent a correlate of protection in chronic HIV-1 infection that is directly relevant to the design of T cell-based vaccines, adoptive immunotherapy approaches, or the pharmacologic induction of T(SCM).


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Immunologic Memory , Stem Cells/immunology , Adult , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/immunology , Cohort Studies , Cross-Sectional Studies , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Prognosis
14.
AIDS Res Hum Retroviruses ; 30(2): 190-4, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23906381

ABSTRACT

Although it has been suggested that biological differences among HIV-1 subtypes exist, their possible influence on disease progression has not been fully revealed. In particular, the increasing emergence of recombinants stresses the need to characterize disease presentation in persons infected by these diverse HIV-1 forms. We explored this issue among 83 Brazilian subjects infected with either HIV-1 subtype B or recombinant subtype BF, all followed since incident infection in a cohort study. Viral subtypes were assigned by full length sequencing of HIV-1 genomes. We observed that the baseline measures for CD4(+) T cells and viral load did not differ between the groups. However, longitudinal analysis revealed that subtype BF was clearly associated with a faster CD4(+) T cell decline compared to infection with subtype B, in spite of a similar plasma HIV-1 load. While subtype B-infected subjects presented a loss of 3.6 CD4(+) T cells/µl per month, subtype BF-infected individuals showed a monthly decay of 6.3 CD4(+) T cells/µl (p<0.01). The time to reach 350 CD4(+) T cells/µl and the time to start antiretroviral treatment were also shorter in subtype BF-infected persons. The elucidation of an accelerated CD4(+) T cell loss associated with subtype BF suggests that this HIV-1 genetic form could be more pathogenic than subtype B.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/classification , HIV-1/isolation & purification , Adult , Brazil , CD4 Lymphocyte Count , Cohort Studies , Female , Genome, Viral , Genotype , HIV-1/genetics , HIV-1/pathogenicity , Humans , Longitudinal Studies , Male , Middle Aged , Sequence Analysis, DNA , Viral Load , Young Adult
15.
J Infect Dis ; 198(4): 614-23, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18598198

ABSTRACT

BACKGROUND: Trypanosoma cruzi-infected outbred hamsters reproduce the range of different outcomes of Chagas disease noted in humans. We tested whether myocarditis, its mediators, and myocardial protein expression are related to the severity of the acute phase of T. cruzi infection in the hamster model. METHODS: Myocardium left ventricles (LVs) obtained from Syrian hamsters infected with T. cruzi were collected 21 days after infection. Myocarditis and the T. cruzi nest/antigen area were analyzed by histological and morphometric analysis. Cytokine and chemokine messenger RNA (mRNA) expression was analyzed using real-time reverse-transcriptase polymerase chain reaction. Differentially expressed proteins were identified by 2-dimensional electrophoresis, followed by mass spectrometry. RESULTS: While in the acute phase of infection, 50% of animals displayed weight loss and signs of acute-phase infection (hereafter referred to as "acute-phase signs" [APS]) (e.g., lethargy, vomiting, and diarrhea). Both the T. cruzi nest/antigen area and the expression of interferon-gamma, tumor necrosis factor-alpha, interleukin-10, and CCL3 mRNA were significantly increased in the LVs of animals with APS, compared with the LVs of animals without APS. Animals with APS, those without APS, and uninfected animals demonstrated distinct myocardial expression of contractile, stress response, and metabolism proteins. CONCLUSIONS: The distinct outcomes of acute T. cruzi infection in Syrian hamsters are related to cardiac parasitism, cytokine expression, and changes in the expression of structural/contractile and stress response proteins that may be associated with alterations in the cardiomyocyte cytoskeleton.


Subject(s)
Cardiomyopathies/immunology , Chagas Cardiomyopathy , Chagas Disease/metabolism , Chemokines/metabolism , Cytokines/metabolism , Trypanosoma cruzi/immunology , Animals , Cardiomyopathies/parasitology , Cardiomyopathies/pathology , Chagas Cardiomyopathy/blood , Chagas Cardiomyopathy/physiopathology , Chagas Disease/physiopathology , Chemokines/genetics , Cricetinae , Cytokines/genetics , Gene Expression , Mice , Myocarditis/blood , Myocarditis/parasitology , Myocarditis/physiopathology , Trypanosoma cruzi/metabolism
16.
Chem Biodivers ; 1(6): 830-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-17191883

ABSTRACT

A novel chemical subclass of toxin, [1-(3-diazenylphenyl)ethanol]iron, was identified among the compounds present in the web of the spider Nephila clavipes. This type of compound is not common among natural products, mainly in spider-venom toxins; it was shown to be a potent paralytic and/or lethal toxin applied by the spider over its web to ensure prey capture only by topical application. The structure was elucidated by means of ESI mass spectrometry, 1H-NMR spectroscopy, high-resolution (HR) mass spectrometry, and ICP spectrometry. The structure of [1-(3-diazenylphenyl)ethanol]iron and the study of its insecticidal action may be used as a starting point for the development of new drugs for pest control in agriculture.


Subject(s)
Ethanol/chemistry , Ferric Compounds/chemistry , Spider Venoms/chemistry , Spider Venoms/classification , Spiders/chemistry , Animals , Ethanol/isolation & purification , Ferric Compounds/classification , Ferric Compounds/isolation & purification , Spider Venoms/isolation & purification , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...