Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 2899, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34006838

RESUMO

There is urgent need for new drug regimens that more rapidly cure tuberculosis (TB). Existing TB drugs and regimens vary in treatment-shortening activity, but the molecular basis of these differences is unclear, and no existing assay directly quantifies the ability of a drug or regimen to shorten treatment. Here, we show that drugs historically classified as sterilizing and non-sterilizing have distinct impacts on a fundamental aspect of Mycobacterium tuberculosis physiology: ribosomal RNA (rRNA) synthesis. In culture, in mice, and in human studies, measurement of precursor rRNA reveals that sterilizing drugs and highly effective drug regimens profoundly suppress M. tuberculosis rRNA synthesis, whereas non-sterilizing drugs and weaker regimens do not. The rRNA synthesis ratio provides a readout of drug effect that is orthogonal to traditional measures of bacterial burden. We propose that this metric of drug activity may accelerate the development of shorter TB regimens.


Assuntos
Antituberculosos/administração & dosagem , Mycobacterium tuberculosis/efeitos dos fármacos , Precursores de RNA/metabolismo , RNA Ribossômico/metabolismo , Tuberculose/tratamento farmacológico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/fisiologia , Precursores de RNA/genética , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA Ribossômico/genética , Resultado do Tratamento , Tuberculose/diagnóstico , Tuberculose/microbiologia
2.
Viruses ; 11(3)2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30871222

RESUMO

Although antiretroviral therapy (ART) greatly suppresses HIV replication, lymphoid tissues remain a sanctuary site where the virus may replicate. Tracking the earliest steps of HIV spread from these cellular reservoirs after drug cessation is pivotal for elucidating how infection can be prevented. In this study, we developed an in vivo model of HIV persistence in which viral replication in the lymphoid compartments of humanized mice was inhibited by the HIV reverse transcriptase inhibitor 4'-ethynyl-2-fluoro-2'-deoxyadenosine (EFdA) to very low levels, which recapitulated ART-suppression in HIV-infected individuals. Using a combination of RNAscope in situ hybridization (ISH) and immunohistochemistry (IHC), we quantitatively investigated the distribution of HIV in the lymphoid tissues of humanized mice during active infection, EFdA suppression, and after drug cessation. The lymphoid compartments of EFdA-suppressed humanized mice harbored very rare transcription/translation-competent HIV reservoirs that enable viral rebound. Our data provided the visualization and direct measurement of the early steps of HIV reservoir expansion within anatomically intact lymphoid tissues soon after EFdA cessation and suggest a strategy to enhance therapeutic approaches aimed at eliminating the HIV reservoir.


Assuntos
Desoxiadenosinas/farmacologia , Reservatórios de Doenças/virologia , HIV-1/fisiologia , Tecido Linfoide/virologia , Inibidores da Transcriptase Reversa/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Feminino , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos
3.
AIDS Res Hum Retroviruses ; 34(1): 3-8, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28691499

RESUMO

The persistence of HIV infection, even after lengthy and successful combined antiretroviral therapy (cART), has precluded an effective cure. The anatomical locations and biological mechanisms through which the viral population is maintained remain unknown. Much research has focused nearly exclusively on circulating resting T cells as the predominant source of persistent HIV, a strategy with limited success in developing an effective cure strategy. In this study, we review research supporting the importance of anatomical tissues and other immune cells for HIV maintenance and expansion, including the central nervous system, lymph nodes, and macrophages. We present accumulated research that clearly demonstrates the limitations of using blood-derived cells as a proxy for tissue reservoirs and sanctuaries throughout the body. We cite recent studies that have successfully used deep-sequencing strategies to uncover the complexity of HIV infection and the ability of the virus to evolve despite undetectable plasma viral loads. Finally, we suggest new strategies and highlight the importance of tissue banks for future research.


Assuntos
Infecções por HIV/tratamento farmacológico , Carga Viral , Latência Viral , Fármacos Anti-HIV/uso terapêutico , Antirretrovirais/uso terapêutico , Terapia Antirretroviral de Alta Atividade , Sistema Nervoso Central/virologia , Infecções por HIV/líquido cefalorraquidiano , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Linfonodos/virologia , Macrófagos/virologia , RNA Viral/sangue , Bancos de Tecidos
4.
PLoS Pathog ; 13(2): e1006202, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28241080

RESUMO

Although invasive cytomegalovirus (CMV) disease is uncommon in the era of antiretroviral therapy (ART), asymptomatic CMV coinfection is nearly ubiquitous in HIV infected individuals. While microbial translocation and gut epithelial barrier dysfunction may promote persistent immune activation in treated HIV infection, potentially contributing to morbidity and mortality, it has been unclear whether CMV replication in individuals with no symptoms of CMV disease might play a role in this process. We hypothesized that persistent CMV replication in the intestinal epithelium of HIV/CMV-coinfected individuals impairs gut epithelial barrier function. Using a combination of state-of-the-art in situ hybridization technology (RNAscope) and immunohistochemistry, we detected CMV DNA and proteins and evidence of intestinal damage in rectosigmoid samples from CMV-positive individuals with both untreated and ART-suppressed HIV infection. Two different model systems, primary human intestinal cells differentiated in vitro to form polarized monolayers and a humanized mouse model of human gut, together demonstrated that intestinal epithelial cells are fully permissive to CMV replication. Independent of HIV, CMV disrupted tight junctions of polarized intestinal cells, significantly reducing transepithelial electrical resistance, a measure of monolayer integrity, and enhancing transepithelial permeability. The effect of CMV infection on the intestinal epithelium is mediated, at least in part, by the CMV-induced proinflammatory cytokine IL-6. Furthermore, letermovir, a novel anti-CMV drug, dampened the effects of CMV on the epithelium. Together, our data strongly suggest that CMV can disrupt epithelial junctions, leading to bacterial translocation and chronic inflammation in the gut and that CMV could serve as a target for therapeutic intervention to prevent or treat gut epithelial barrier dysfunction during HIV infection.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus , Infecções por HIV/virologia , Mucosa Intestinal/virologia , Replicação Viral , Animais , Coinfecção , Infecções por Citomegalovirus/imunologia , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Interleucina-6/biossíntese , Interleucina-6/imunologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos SCID , Permeabilidade
5.
J Virol ; 90(20): 8984-93, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27466425

RESUMO

UNLABELLED: While combined antiretroviral therapy (cART) can result in undetectable plasma viral loads, it does not eradicate HIV infection. Furthermore, HIV-infected individuals while on cART remain at an increased risk of developing serious comorbidities, such as cancer, neurological disease, and atherosclerosis, suggesting that during cART, tissue-based HIV may contribute to such pathologies. We obtained DNA and RNA env, nef, and pol sequences using single-genome sequencing from postmortem tissues of three HIV(+) cART-treated (cART(+)) individuals with undetectable viral load and metastatic cancer at death and performed time-scaled Bayesian evolutionary analyses. We used a sensitive in situ hybridization technique to visualize HIV gag-pol mRNA transcripts in cerebellum and lymph node tissues from one patient. Tissue-associated virus evolved at similar rates in cART(+) and cART-naive (cART(-)) patients. Phylogenetic trees were characterized by two distinct features: (i) branching patterns consistent with constant viral evolution and dispersal among tissues and (ii) very recently derived clades containing both DNA and RNA sequences from multiple tissues. Rapid expansion of virus near death corresponded to wide-spread metastasis. HIV RNA(+) cells clustered in cerebellum tissue but were dispersed in lymph node tissue, mirroring the evolutionary patterns observed for that patient. Activated, infiltrating macrophages were associated with HIV RNA. Our data provide evidence that tissues serve as a sanctuary for wild-type HIV during cART and suggest the importance of macrophages as an alternative reservoir and mechanism of virus spread. IMPORTANCE: Combined antiretroviral therapy (cART) reduces plasma HIV to undetectable levels; however, removal of cART results in plasma HIV rebound, thus highlighting its inability to entirely rid the body of infection. Additionally, HIV-infected individuals on cART remain at high risk of serious diseases, which suggests a contribution from residual HIV. In this study, we isolated and sequenced HIV from postmortem tissues from three HIV(+) cART(+) individuals who died with metastatic cancer and had no detectable plasma viral load. Using high-resolution evolutionary analyses, we found that tissue-based HIV continues to replicate, evolve, and migrate among tissues during cART. Furthermore, cancer onset and metastasis coincided with increased HIV expansion, suggesting a linked mechanism. HIV-expressing cells were associated with tissue macrophages, a target of HIV infection. Our results suggest the importance of tissues, and macrophages in particular, as a target for novel anti-HIV therapies.


Assuntos
Antirretrovirais/uso terapêutico , Infecções por HIV/complicações , Infecções por HIV/virologia , HIV/isolamento & purificação , Neoplasias/complicações , Resposta Viral Sustentada , Carga Viral , Terapia Antirretroviral de Alta Atividade , Autopsia , Cerebelo/virologia , DNA Viral/genética , Variação Genética , HIV/classificação , HIV/genética , Infecções por HIV/tratamento farmacológico , Hibridização In Situ , Linfonodos/virologia , Filogenia , RNA Viral/genética , Análise de Sequência de DNA , Homologia de Sequência , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene pol do Vírus da Imunodeficiência Humana/genética
6.
J Virol ; 90(20): 8968-83, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27466426

RESUMO

UNLABELLED: HIV infection treatment strategies have historically defined effectiveness through measuring patient plasma HIV RNA. While combined antiretroviral therapy (cART) can reduce plasma viral load (pVL) to undetectable levels, the degree that HIV is eliminated from other anatomical sites remains unclear. We investigated the HIV DNA levels in 229 varied autopsy tissues from 20 HIV-positive (HIV(+)) cART-treated study participants with low or undetectable plasma VL and cerebrospinal fluid (CSF) VL prior to death who were enrolled in the National Neurological AIDS Bank (NNAB) longitudinal study and autopsy cohort. Extensive medical histories were obtained for each participant. Autopsy specimens, including at least six brain and nonbrain tissues per participant, were reviewed by study pathologists. HIV DNA, measured in tissues by quantitative and droplet digital PCR, was identified in 48/87 brain tissues and 82/142 nonbrain tissues at levels >200 HIV copies/million cell equivalents. No participant was found to be completely free of tissue HIV. Parallel sequencing studies from some tissues recovered intact HIV DNA and RNA. Abnormal histological findings were identified in all participants, especially in brain, spleen, lung, lymph node, liver, aorta, and kidney. All brain tissues demonstrated some degree of pathology. Ninety-five percent of participants had some degree of atherosclerosis, and 75% of participants died with cancer. This study assists in characterizing the anatomical locations of HIV, in particular, macrophage-rich tissues, such as the central nervous system (CNS) and testis. Additional studies are needed to determine if the HIV recovered from tissues promotes the pathogenesis of inflammatory diseases, such as HIV-associated neurocognitive disorders, cancer, and atherosclerosis. IMPORTANCE: It is well-known that combined antiretroviral therapy (cART) can reduce plasma HIV to undetectable levels; however, cART cannot completely clear HIV infection. An ongoing question is, "Where is HIV hiding?" A well-studied HIV reservoir is "resting" T cells, which can be isolated from blood products and succumb to cART once activated. Less-studied reservoirs are anatomical tissue samples, which have unknown cART penetration, contain a comparably diverse spectrum of potentially HIV-infected immune cells, and are important since <2% of body lymphocytes actually reside in blood. We examined 229 varied autopsy specimens from 20 HIV(+) participants who died while on cART and identified that >50% of tissues were HIV infected. Additionally, we identified considerable pathology in participants' tissues, especially in brain, spleen, lung, lymph node, liver, aorta, and kidney. This study substantiates that tissue-associated HIV is present despite cART and can inform future studies into HIV persistence.


Assuntos
Antirretrovirais/uso terapêutico , Autopsia , DNA Viral/análise , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Carga Viral , Humanos , Estudos Longitudinais , Reação em Cadeia da Polimerase em Tempo Real
7.
J Biol Chem ; 291(19): 10332-46, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-26957545

RESUMO

HIV evades eradication because transcriptionally dormant proviral genomes persist in long-lived reservoirs of resting CD4(+) T cells and myeloid cells, which are the source of viral rebound after cessation of antiretroviral therapy. Dormant HIV genomes readily produce infectious virus upon cellular activation because host transcription factors activated specifically by cell stress and heat shock mediate full-length HIV transcription. The molecular chaperone heat shock protein 90 (Hsp90) is overexpressed during heat shock and activates inducible cellular transcription factors. Here we show that heat shock accelerates HIV transcription through induction of Hsp90 activity, which activates essential HIV-specific cellular transcription factors (NF-κB, NFAT, and STAT5), and that inhibition of Hsp90 greatly reduces gene expression mediated by these factors. More importantly, we show that Hsp90 controls virus transcription in vivo by specific Hsp90 inhibitors in clinical development, tanespimycin (17-(allylamino)-17-demethoxygeldanamycin) and AUY922, which durably prevented viral rebound in HIV-infected humanized NOD scid IL-2Rγ(-/-) bone marrow-liver-thymus mice up to 11 weeks after treatment cessation. Despite the absence of rebound viremia, we were able to recover infectious HIV from PBMC with heat shock. Replication-competent virus was detected in spleen cells from these nonviremic Hsp90 inhibitor-treated mice, indicating the presence of a tissue reservoir of persistent infection. Our novel findings provide in vivo evidence that inhibition of Hsp90 activity prevents HIV gene expression in replication-competent cellular reservoirs that would typically cause rebound in plasma viremia after antiretroviral therapy cessation. Alternating or supplementing Hsp90 inhibitors with current antiretroviral therapy regimens could conceivably suppress rebound viremia from persistent HIV reservoirs.


Assuntos
Infecções por HIV/prevenção & controle , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Latência Viral/efeitos dos fármacos , Animais , Antivirais/farmacologia , Benzoquinonas/farmacologia , Western Blotting , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Feminino , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Lactamas Macrocíclicas/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
8.
J Neurovirol ; 22(3): 275-81, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26572785

RESUMO

Two innovative studies recently identified functional lymphatic structures in the meninges that may influence the development of HIV-associated neurological disorders (HAND). Until now, blood vessels were assumed to be the sole transport system by which HIV-infected monocytes entered the brain by bypassing a potentially hostile blood-brain barrier through inflammatory-mediated semi-permeability. A cascade of specific chemokine signals promote monocyte migration from blood vessels to surrounding brain tissues via a well-supported endothelium, where the cells differentiate into tissue macrophages capable of productive HIV infection. Lymphatic vessels on the other hand are more loosely organized than blood vessels. They absorb interstitial fluid from bodily tissues where HIV may persist and exchange a variety of immune cells (CD4(+) T cells, monocytes, macrophages, and dendritic cells) with surrounding tissues through discontinuous endothelial junctions. We propose that the newly discovered meningeal lymphatics are key to HIV migration among viral reservoirs and brain tissue during periods of undetectable plasma viral loads due to suppressive combinational antiretroviral therapy, thus redefining the migration process in terms of a blood-lymphatic transport system.


Assuntos
Complexo AIDS Demência/virologia , Encéfalo/virologia , HIV-1/fisiologia , Sistema Linfático/virologia , Meninges/virologia , Monócitos/virologia , Complexo AIDS Demência/imunologia , Complexo AIDS Demência/patologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/virologia , Encéfalo/imunologia , Movimento Celular , Quimiocinas/biossíntese , Quimiocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/virologia , Endotélio Vascular/imunologia , Endotélio Vascular/virologia , HIV-1/patogenicidade , Humanos , Sistema Linfático/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Meninges/imunologia , Monócitos/imunologia , Linfócitos T/imunologia , Linfócitos T/virologia , Internalização do Vírus
9.
Virology ; 462-463: 115-25, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24971704

RESUMO

Highly potent broadly neutralizing human monoclonal antibodies hold promise for HIV prophylaxis and treatment. We used the SCID-hu Thy/Liv and BLT humanized mouse models to study the efficacy of these antibodies, primarily PG16, against HIV-1 clades A, B, and C. PG16 targets a conserved epitope in the V1/V2 region of gp120 common to 70-80% of HIV-1 isolates from multiple clades and has extremely potent in vitro activity against HIVJR-CSF. PG16 was highly efficacious in SCID-hu mice as a single intraperitoneal administration the day before inoculation of R5-tropic HIV directly into their Thy/Liv implants and demonstrated even greater efficacy if PG16 administration was continued after Thy/Liv implant HIV inoculation. However, PG16 as monotherapy had no activity in humanized mice with established R5-tropic HIV infection. These results provide evidence of tissue penetration of the antibodies, which could aid in their ability to prevent infection if virus crosses the mucosal barrier.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Anti-HIV/uso terapêutico , Infecções por HIV/terapia , HIV-1/imunologia , Animais , Modelos Animais de Doenças , Proteína gp120 do Envelope de HIV/imunologia , Masculino , Camundongos , Camundongos SCID , Resultado do Tratamento
10.
J Infect Dis ; 209(10): 1573-84, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24403553

RESUMO

BACKGROUND: Human cytomegalovirus (HCMV) is the major viral etiology of congenital infection and birth defects. Fetal transmission is high (30%-40%) in primary maternal infection, and symptomatic babies have permanent neurological, hearing, and vision defects. Recurrent infection is infrequently transmitted (2%) and largely asymptomatic. Congenital infection is also associated with intrauterine growth restriction (IUGR). METHODS: To investigate possible underlying HCMV infection in cases of idiopathic IUGR, we studied maternal and cord sera and placentas from 19 pregnancies. Anti-HCMV antibodies, hypoxia-related factors, and cmvIL-10 were measured in sera. Placental biopsy specimens were examined for viral DNA, expression of infected cell proteins, and pathology. RESULTS: Among 7 IUGR cases, we identified 2 primary and 3 recurrent HCMV infections. Virus replicated in glandular epithelium and lymphatic endothelium in the decidua, cytotrophoblasts, and smooth muscle cells in blood vessels of floating villi and the chorion. Large fibrinoids with avascular villi, edema, and inflammation were significantly increased. Detection of viral proteins in the amniotic epithelium indicated transmission in 2 cases of IUGR with primary infection and 3 asymptomatic recurrent infections. CONCLUSIONS: Congenital HCMV infection impairs placental development and functions and should be considered as an underlying cause of IUGR, regardless of virus transmission to the fetus.


Assuntos
Infecções por Citomegalovirus/complicações , Retardo do Crescimento Fetal/virologia , Complicações Infecciosas na Gravidez/patologia , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , DNA Viral , Feminino , Humanos , Imunoglobulina G/sangue , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Projetos Piloto , Gravidez , Testes Sorológicos
11.
J Virol ; 86(23): 12795-805, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22973041

RESUMO

Human cytomegalovirus (HCMV) is the leading viral cause of birth defects and life-threatening lung-associated diseases in premature infants and immunocompromised children. Although the fetal lung is a major target organ of the virus, HCMV lung pathogenesis has remained unexplored, possibly as a result of extreme host range restriction. To overcome this hurdle, we generated a SCID-hu lung mouse model that closely recapitulates the discrete stages of human lung development in utero. Human fetal lung tissue was implanted into severe combined immunodeficient (CB17-scid) mice and inoculated by direct injection with the VR1814 clinical isolate of HCMV. Virus replication in the fetal lung was assessed by the quantification of infectious virus titers and HCMV genome copies and the detection of HCMV proteins by immunohistochemistry and Western blotting. We show that HCMV efficiently replicated in the lung implants during a 2-week period, forming large viral lesions. The virus productively infected alveolar epithelial and mesenchymal cells, imitating congenital infection of the fetal lung. HCMV replication triggered apoptosis near and within the viral lesions and impaired the production of surfactant proteins in the alveolar epithelium. Our findings highlight that congenital and neonatal HCMV infection can adversely impact lung development, leading to pneumonia and acute lung injury. We have successfully developed a small-animal model that closely recapitulates fetal and neonatal lung development and provides a valuable, biologically relevant tool for an understanding of the lung pathogenesis of HCMV as well as other human respiratory viruses. Additionally, this model would greatly facilitate the development and testing of new antiviral therapies for HCMV along with select human pulmonary pathogens.


Assuntos
Infecções por Citomegalovirus/congênito , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Alvéolos Pulmonares/virologia , Proteínas Associadas a Surfactantes Pulmonares/metabolismo , Replicação Viral/fisiologia , Animais , Apoptose/fisiologia , Western Blotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos SCID , Alvéolos Pulmonares/citologia , Estatísticas não Paramétricas , Proteínas Virais/isolamento & purificação
12.
AIDS ; 26(7): 805-14, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22313962

RESUMO

OBJECTIVE: HIV-infected individuals are at increased risk for myocardial infarction. Given observations that cytomegalovirus (CMV) infection, CMV-specific T cells, and CX3CR1 have each been associated with atherosclerosis, we hypothesized that CMV-induced T-cell immunopathology could contribute to HIV-associated atherosclerosis. METHODS: We measured the expression of CX3CR1 on peripheral blood mononuclear cells and its association with carotid artery intima-media thickness (IMT) in 29 HIV-infected individuals and 48 uninfected controls. We analyzed the phenotype and specificity of CX3CR1(+)CD4(+) T cells, the production of CX3CL1 (the ligand of CX3CR1) by CMV-infected endothelial cells in vitro, and the migration of CD4(+) T cells induced by CX3CL1. RESULTS: The progression of atherosclerosis in HIV-infected individuals, as assessed by longitudinal measurements of carotid IMT, was associated with a high frequency of CD4(+) T cells that express the chemokine receptor CX3CR1. Such CD4(+)CX3CR1(+) T cells were antigen-primed, produced high levels of pro-inflammatory cytokines, and composed the majority of the CMV-specific CD4(+) T cells. CMV-stimulated CD4(+) T cells were also found to induce the production of CX3CL1 (the ligand for CX3CR1) by human arterial endothelial cells, driving the transendothelial migration of pro-inflammatory CD4(+) T cells. Finally, we observed that CD4(+)CX3CR1(+) T cells could be localized to the coronary arterial wall in HIV disease. CONCLUSION: HIV-associated atherosclerosis may be driven by a positive feedback pathway in which a high frequency of antigen-stimulated, CMV-specific CD4(+)CX3CR1(+) T cells induce endothelial cells to secrete CX3CL1, which itself drives progressive infiltration of the arterial wall by pro-inflammatory cells.


Assuntos
Aterosclerose/imunologia , Linfócitos T CD4-Positivos/imunologia , Espessura Intima-Media Carotídea , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Infecções por HIV/imunologia , Adulto , Aterosclerose/complicações , Linfócitos T CD4-Positivos/metabolismo , Receptor 1 de Quimiocina CX3C , Estudos de Casos e Controles , Quimiocina CX3CL1/metabolismo , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/complicações , Células Endoteliais/patologia , Feminino , Infecções por HIV/complicações , Humanos , Masculino , Pessoa de Meia-Idade , Receptores de Quimiocinas/metabolismo
13.
Virology ; 417(1): 154-60, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21684569

RESUMO

Humanized Bone marrow/Liver/Thymus (BLT) mice recapitulate the mucosal transmission of HIV, permitting study of early events in HIV pathogenesis and evaluation of preexposure prophylaxis methods to inhibit HIV transmission. Human hematopoiesis is reconstituted in NOD-scid mice by implantation of human fetal liver and thymus tissue to generate human T cells plus intravenous injection of autologous liver-derived CD34(+) hematopoietic stem cells to engraft the mouse bone marrow. In side-by-side comparisons, we show that NOD-scid mice homozygous for a deletion of the IL-2Rγ-chain (NOD-scid IL-2Rγ(-/-)) are far superior to NOD-scid mice in both their peripheral blood reconstitution with multiple classes of human leukocytes (e.g., a mean of 182 versus 14 CD4(+) T cells per µl 12 weeks after CD34(+) injection) and their susceptibility to intravaginal HIV exposure (84% versus 11% viremic mice at 4 weeks). These results should speed efforts to obtain preclinical animal efficacy data for new HIV drugs and microbicides.


Assuntos
Infecções por HIV/transmissão , HIV/imunologia , Subunidade gama Comum de Receptores de Interleucina/fisiologia , Leucócitos/fisiologia , Animais , Antígenos CD34 , Suscetibilidade a Doenças , Feminino , HIV/fisiologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Transplante Heterólogo , Vagina
14.
Am J Pathol ; 177(3): 1298-310, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20651234

RESUMO

Human cytomegalovirus (HCMV) is the major viral cause of birth defects worldwide. Affected infants can have temporary symptoms that resolve soon after birth, such as growth restriction, and permanent disabilities, including neurological impairment. Passive immunization of pregnant women with primary HCMV infection is a promising treatment to prevent congenital disease. To understand the effects of sustained viral replication on the placenta and passive transfer of protective antibodies, we performed immunohistological analysis of placental specimens from women with untreated congenital infection, HCMV-specific hyperimmune globulin treatment, and uninfected controls. In untreated infection, viral replication proteins were found in trophoblasts and endothelial cells of chorionic villi and uterine arteries. Associated damage included extensive fibrinoid deposits, fibrosis, avascular villi, and edema, which could impair placental functions. Vascular endothelial growth factor and its receptor fms-like tyrosine kinase 1 (Flt1) were up-regulated, and amniotic fluid contained elevated levels of soluble Flt1 (sFlt1), an antiangiogenic protein, relative to placental growth factor. With hyperimmune globulin treatment, placentas appeared uninfected, vascular endothelial growth factor and Flt1 expression was reduced, and sFlt1 levels in amniotic fluid were lower. An increase in the number of chorionic villi and blood vessels over that in controls suggested compensatory development for a hypoxia-like condition. Taken together the results indicate that antibody treatment can suppress HCMV replication and prevent placental dysfunction, thus improving fetal outcome.


Assuntos
Vilosidades Coriônicas/virologia , Infecções por Citomegalovirus/metabolismo , Placenta/virologia , Trofoblastos/virologia , Proteínas Virais/metabolismo , Análise de Variância , Vilosidades Coriônicas/metabolismo , Vilosidades Coriônicas/patologia , Citomegalovirus , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Placenta/metabolismo , Placenta/patologia , Gravidez , Trofoblastos/metabolismo , Trofoblastos/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
J Clin Virol ; 46 Suppl 4: S58-63, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19854676

RESUMO

BACKGROUND: Cytomegalovirus (CMV) is the major cause of congenital infection and disease leading to permanent birth defects. In about 35-40% of pregnant women with primary CMV infection, virus crosses the placenta, resulting in the birth of congenitally infected babies. In contrast, this happens in only 1-3% of seropositive women with strong CMV-specific humoral immunity. Whether CMV reaches the fetus and disseminates depends on the level of high-avidity antibodies in the maternal circulation and the passive immunity of the fetus. OBJECTIVES AND STUDY DESIGN: To identify CMV infection in uncomplicated deliveries based on detection of viral DNA in placental biopsy specimens at term. To quantify CMV-specific IgG avidity, neutralizing titer, IgG1 concentration, and characterize the immunoblot profiles for CMV proteins in paired samples of placental and cord blood sera. RESULTS: In accord with earlier reports, CMV DNA was detected in 39% (11/28) of placentas with mean- to high-avidity CMV-specific IgG. In seropositive women, the concentration of antiviral antibodies, specifically IgG1, increased in the fetal bloodstream, and CMV neutralizing titers in maternal and fetal blood were comparable. CONCLUSIONS: CMV-specific, high-avidity neutralizing antibodies from maternal circulation are transcytosed to the fetal bloodstream, contribute to suppression of viral replication in the placenta and could prevent congenital disease.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Feto/imunologia , Imunoglobulina G/sangue , Complicações Infecciosas na Gravidez/imunologia , Afinidade de Anticorpos/imunologia , Infecções por Citomegalovirus/sangue , Feminino , Sangue Fetal/imunologia , Sangue Fetal/virologia , Feto/virologia , Humanos , Transmissão Vertical de Doenças Infecciosas , Testes de Neutralização , Placenta/imunologia , Placenta/virologia , Gravidez , Complicações Infecciosas na Gravidez/sangue , Complicações Infecciosas na Gravidez/virologia
16.
Am J Pathol ; 172(4): 1127-40, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18349127

RESUMO

Human cytomegalovirus (CMV) infection is a major cause of morbidity in immunosuppressed individuals, and congenital CMV infection is a leading cause of birth defects in newborns. Infection with pathogenic viral strains alters cell-cell and cell-matrix interactions, affecting extracellular matrix remodeling and endothelial cell migration. The multifunctional cytokine transforming growth factor (TGF)-beta1 regulates cell proliferation, differentiation, and extracellular matrix remodeling. Secreted as a latent protein complex, TGF-beta1 requires activation before binding to receptors that phosphorylate intracellular effectors. TGF-beta1 is activated by integrin alphavbeta6, which is strongly induced in the epithelium by injury and inflammation but has not previously been found in endothelial cells. Here, we report that CMV infection induces integrin alphavbeta6 expression in endothelial cells, leading to activation of TGF-beta1, signaling through its receptor ALK5, and phosphorylation of its intracellular effector Smad3. Infection of endothelial cells was also found to stimulate collagen synthesis through a mechanism dependent on both TGF-beta1 and integrin alphavbeta6. Immunohistochemical analysis showed integrin alphavbeta6 up-regulation in capillaries proximal to foci of CMV infection in lungs, salivary glands, uterine decidua, and injured chorionic villi of the placenta, demonstrating both its induction in endothelium and up-regulation in epithelium in vivo. Our results suggest that activation of TGF-beta1 by integrin alphavbeta6 contributes to pathological changes and may impair endothelial cell functions in tissues that are chronically infected with CMV.


Assuntos
Antígenos de Neoplasias/metabolismo , Colágeno/biossíntese , Infecções por Citomegalovirus/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Células Epiteliais/metabolismo , Integrinas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Células Cultivadas , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Células Endoteliais/patologia , Matriz Extracelular/metabolismo , Humanos , Cadeias beta de Integrinas/metabolismo , Vison , Especificidade de Órgãos , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Veias Umbilicais/patologia , Veias Umbilicais/virologia , Regulação para Cima , Replicação Viral
17.
J Virol ; 81(9): 4701-12, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17314173

RESUMO

Cytomegalovirus (CMV), the major viral cause of congenital disease, infects the uterus and developing placenta and spreads to the fetus throughout gestation. Virus replicates in invasive cytotrophoblasts in the decidua, and maternal immunoglobulin G (IgG)-CMV virion complexes, which are transcytosed by the neonatal Fc receptor across syncytiotrophoblasts, infect underlying cytotrophoblasts in chorionic villi. Immunity is central to protection of the placenta-fetal unit: infection can occur when IgG has a low neutralizing titer. Here we used immunohistochemical and function-blocking methods to correlate infection in the placenta with expression of potential CMV receptors in situ and in vitro. In placental villi, syncytiotrophoblasts express the virion receptor epidermal growth factor receptor (EGFR) but lack integrin coreceptors, and virion uptake occurs without replication. Focal infection can occur when transcytosed virions reach EGFR-expressing cytotrophoblasts that selectively initiate expression of alphaV integrin. In cell columns, proximal cytotrophoblasts lack receptors and distal cells express integrins alpha1beta1 and alphaVbeta3, enabling virion attachment. In the decidua, invasive cytotrophoblasts expressing coreceptors upregulate EGFR, thereby dramatically increasing susceptibility to infection. Our findings indicate that virion interactions with cytotrophoblasts expressing receptors in the placenta (i) change as the cells differentiate and (ii) correlate with spatially distinct sites of CMV replication in maternal and fetal compartments.


Assuntos
Infecções por Citomegalovirus/metabolismo , Regulação da Expressão Gênica , Placenta/citologia , Receptores Virais/metabolismo , Trofoblastos/metabolismo , Replicação Viral/fisiologia , Receptores ErbB/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Integrinas/metabolismo , Microscopia de Fluorescência , Gravidez , Trofoblastos/virologia , Ligação Viral
18.
Am J Pathol ; 168(4): 1210-26, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16565496

RESUMO

How human cytomegalovirus (CMV) reaches the fetus across the placenta is unknown. The major viral cause of congenital disease, CMV infects the uterine-placental interface with varied outcomes depending on the strength of maternal humoral immunity and gestational age. Covering the surface of villi that float in blood, syncytiotrophoblasts express the neonatal Fc receptor (FcRn) that transports IgG for passive immunity. Immunohistochemical analysis of early-gestation biopsy specimens showed an unusual pattern of CMV replication proteins in underlying villus cytotrophoblasts, whereas syncytiotrophoblasts were spared. Found in placentas with low to moderate CMV-neutralizing antibody titers, this pattern suggested virion transcytosis across the surface. In contrast, syncytiotrophoblasts from placentas with high neutralizing titers contained viral DNA and caveolin-1-positive vesicles in which IgG and CMV glycoprotein B co-localized. In villus explants, IgG-virion transcytosis and macrophage uptake were blocked with trypsin-treatment and soluble protein A. Quantitative analysis in polarized epithelial cells showed that FcRn-mediated transcytosis was blocked by the Fc fragment of IgG, but not F(ab')(2). Our results suggest that CMV virions could disseminate to the placenta by co-opting the receptor-mediated transport pathway for IgG. These findings could explain the efficacy of hyperimmune IgG for treatment of primary CMV infection during gestation and support vaccination.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunidade Materno-Adquirida , Fragmentos Fc das Imunoglobulinas/imunologia , Placenta/imunologia , Complicações Infecciosas na Gravidez/imunologia , Receptores Fc/imunologia , Linhagem Celular Tumoral , Vilosidades Coriônicas/imunologia , Vilosidades Coriônicas/virologia , Infecções por Citomegalovirus/virologia , DNA Viral/análise , Feminino , Humanos , Imunoglobulina G/imunologia , Macrófagos/imunologia , Técnicas de Cultura de Órgãos , Placenta/virologia , Gravidez , Complicações Infecciosas na Gravidez/virologia , Trofoblastos/imunologia , Trofoblastos/virologia , Útero/virologia , Vírion/fisiologia , Replicação Viral
19.
J Clin Virol ; 35(2): 210-5, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16386950

RESUMO

BACKGROUND: Primary maternal CMV infection is the major risk factor for symptomatic congenital infection as maternal immunity reduces the risk of transmission to the fetus. Analysis of first trimester placentas showed that virus replicates in the uterus and is transmitted to the placenta causing focal infection. OBJECTIVES AND STUDY DESIGN: We examined 78 term placentas from uncomplicated deliveries for the presence of CMV DNA and evaluated evidence of infection by means of immunohistological and serological analysis. RESULTS: PCR analysis of villus biopsy samples and decidua showed that CMV DNA was present in 62% of tissues. Seven placentas with neutralizing titers were further examined by immunohistology for expression of viral proteins. In placentas with high levels of CMV DNA, fetal blood vessels in the villus core contained neutrophils with viral replication proteins, and macrophages/dendritic cells with glycoprotein B (gB). Cord blood samples from 1 of 11 placentas contained CMV DNA, an indication of replication in the fetal compartment. In placentas with low levels of viral DNA, macrophage/dendritic cells in the villus core contained CMV gB. This pattern was comparable to that seen in early gestation placentas from women with strong neutralizing antibodies. CONCLUSIONS: The results show CMV replication proteins in focal areas of the placenta, implying virus transmission to the fetal circulation. These preliminary results suggest that the incidence of asymptomatic congenital CMV infection might be higher than currently estimated.


Assuntos
Infecções por Citomegalovirus/fisiopatologia , Citomegalovirus/isolamento & purificação , Transmissão Vertical de Doenças Infecciosas , Doenças Placentárias/virologia , Complicações Infecciosas na Gravidez , Citomegalovirus/genética , Infecções por Citomegalovirus/transmissão , DNA Viral/análise , Feminino , Humanos , Doenças Placentárias/patologia , Gravidez
20.
Trends Microbiol ; 13(4): 164-74, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15817386

RESUMO

During human gestation, viruses can cause intrauterine infections associated with pregnancy complications and fetal abnormalities. The ability of viruses to spread from the infected mother to the fetus arises from the architecture of the placenta, which anchors the fetus to the uterus. Placental cytotrophoblasts differentiate, assume an endothelial phenotype, breach uterine blood vessels and form a hybrid vasculature that amplifies the maternal blood supply for fetal development. Human cytomegalovirus - the major cause of congenital disease - infects the uterine wall and the adjacent placenta, suggesting adaptation for pathogen survival in this microenvironment. Infection of villus explants and differentiating and/or invading cytotrophoblasts offers an in vitro model for studying viruses associated with prenatal infections.


Assuntos
Infecções por Citomegalovirus/transmissão , Doenças Fetais/virologia , Placenta/virologia , Complicações Infecciosas na Gravidez/virologia , Útero/virologia , Viroses/transmissão , Animais , Infecções por Citomegalovirus/congênito , Infecções por Citomegalovirus/virologia , Feminino , Humanos , Circulação Placentária , Gravidez , Doenças Uterinas/virologia , Viroses/congênito , Viroses/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA