Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pediatr Blood Cancer ; 66(8): e27790, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31045327

RESUMO

BACKGROUND: Kaposiform lymphangiomatosis (KLA) is a rare lymphatic anomaly with significant morbidity and mortality. KLA is characterized by diffuse multifocal lesions comprised of focal areas of "kaposiform" spindled cells accompanying malformed lymphatic channels. The goal of this study was to identify activated signaling pathways in cells isolated from three KLA patients for the purpose of testing new therapies. PROCEDURE: Cells were obtained from the lungs of one patient isolated at autopsy and the spleen of two patients removed in surgery due to disease complications. A protein kinase array was performed on the KLA cell lysates and normal lymphatic endothelial cells. RESULTS: Higher activation of key signaling pathways in the KLA cells, including PRAS40, AKT1/2/3, and ERK-1/2, was identified by protein kinase array and confirmed by Western blot analysis. This indicated a role for highly activated PI3K-AKT and MAPK-ERK-1/2 signaling pathways in KLA cells. Cell proliferation studies assessed PI3K inhibitors (LY294002; BYL719), AKT inhibitor ARQ092, mTOR inhibitor rapamycin, and MAPK inhibitor U0126. These studies demonstrated that PI3K-AKT-mTOR and MAPK signaling are important mediators of KLA cell proliferation. BYL719 and rapamycin were more effective at inhibiting KLA cell proliferation than U0126. CONCLUSIONS: Our studies using cells from KLA patient lesions demonstrate that these cells are highly proliferative and the PI3K-AKT-mTOR and MAPK pathways are promising therapeutic targets. Development and clinical trials of PI3K, AKT, and MAPK inhibitors for cancer treatment and the data in this study lend support for early clinical trials assessing the efficacy of these inhibitors in KLA patients.


Assuntos
Antineoplásicos/farmacologia , Hemangioendotelioma/patologia , Síndrome de Kasabach-Merritt/patologia , Linfangioma/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/química , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Sarcoma de Kaposi/patologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Adolescente , Proliferação de Células/efeitos dos fármacos , Pré-Escolar , Feminino , Seguimentos , Hemangioendotelioma/tratamento farmacológico , Hemangioendotelioma/metabolismo , Humanos , Lactente , Síndrome de Kasabach-Merritt/tratamento farmacológico , Síndrome de Kasabach-Merritt/metabolismo , Linfangioma/tratamento farmacológico , Linfangioma/metabolismo , Masculino , Inibidores de Proteínas Quinases/farmacologia , Estudos Retrospectivos , Sarcoma de Kaposi/tratamento farmacológico , Sarcoma de Kaposi/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
2.
J Pediatr Hematol Oncol ; 40(8): e495-e504, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30256265

RESUMO

Kaposiform lymphangiomatosis (KLA) is a vascular anomaly featuring lymphatic expansion. It has no known cause, no effective treatment, and is associated with high morbidity. Proliferative cells from 3 KLA patient lesions were characterized relative to adiopose-derived mesenchyme stem cells (ADSCs) and cells derived from a patient with the related disease kaposiform hemangioendothelioma (KHE). KLA cells variably expressed markers of mesenchyme stem cells (CD73, CD90, CD105, CD146) and lacked endothelial cell markers (CD31, CD34) as determined by flow cytometry. They expressed markers of vascular pericytes (neural/glial antigen 2, alpha-smooth muscle actin, platelet-derived growth factor-beta receptor, and CXCL12) as determined by quantitative reverse transcription polymerase chain reaction. Lesion cells transcribed vascular markers VEGFC and VEGFD, as well as VCAM-1, the latter of which was confirmed by flow cytometry, consistent with angiogenic MSC-like pericytes. Furthermore, conditioned medium from each was shown to promote the proliferation of growth factor-starved lymphatic endothelial cells. Unlike kaposiform hemangioendothelioma-derived MSC-like pericytes and ADSCs, KLA isolates were defective in support of vascular network formation in co-cultures with either vascular or lymphatic endothelial cells. Genetic analysis by whole exome sequencing revealed novel variant alleles in 2 populations of KLA cells (BAD, TSC1) that may bear on aberrant pericyte growth and function.


Assuntos
Proliferação de Células , Hemangioendotelioma/metabolismo , Síndrome de Kasabach-Merritt/metabolismo , Linfangiectasia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Patológica/metabolismo , Pericitos/metabolismo , Sarcoma de Kaposi/metabolismo , Adolescente , Antígenos de Diferenciação/biossíntese , Criança , Pré-Escolar , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Hemangioendotelioma/patologia , Humanos , Síndrome de Kasabach-Merritt/patologia , Linfangiectasia/patologia , Masculino , Células-Tronco Mesenquimais/patologia , Proteínas de Neoplasias/biossíntese , Neovascularização Patológica/patologia , Pericitos/patologia , Sarcoma de Kaposi/patologia
3.
Hum Mol Genet ; 24(4): 926-38, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25292196

RESUMO

Lymphatic malformations (LMs) are developmental anomalies of the lymphatic system associated with the dysmorphogenesis of vascular channels lined by lymphatic endothelial cells (LECs). Seeking to identify intrinsic defects in affected LECs, cells were isolated from malformation tissue or fluid on the basis of CD31 and podoplanin (PDPN) expression. LECs from five unrelated LM lesions were characterized, including cells derived from one patient previously diagnosed with CLOVES. CLOVES-related LECs carried a known, activating mutation in PIK3CA (p.H1047L), confirmed by direct sequencing. Activating PIK3CA mutations (p.E542K and p.E545A) were identified in lesion-derived cells from the other four patients, also by direct sequencing. The five LM-LEC cultures shared a lymphangiogenic phenotype distinguished by PI3K/AKT activation, enhanced sprouting efficiency, elevated VEGF-C expression and COX2 expression, shorter doubling times and reduced expression of angiopoietin 2 and CXCR4. Nine additional LM-LEC populations and 12 of 15 archived LM tissue samples were shown to bear common PIK3CA variants by allele-specific PCR. The activation of a central growth/survival pathway (PI3K/AKT) represents a feasible target for the non-invasive treatment of LMs bearing in mind that background genetics may individualize lesions and influence treatments.


Assuntos
Alelos , Células Endoteliais/metabolismo , Linfangiogênese/genética , Anormalidades Linfáticas/genética , Anormalidades Linfáticas/patologia , Fenótipo , Fosfatidilinositol 3-Quinases/genética , Ativação Transcricional , Adolescente , Criança , Pré-Escolar , Classe I de Fosfatidilinositol 3-Quinases , Análise Mutacional de DNA , Células Endoteliais/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Humanos , Anormalidades Linfáticas/diagnóstico , Masculino , Mutação , Naftiridinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais , Adulto Jovem
4.
Gastroenterology ; 147(1): 162-171.e6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24662331

RESUMO

BACKGROUND & AIMS: Type 1 diabetes is caused by an aberrant response against pancreatic ß cells. Intestinal K cells are glucose-responsive endocrine cells that might be engineered to secrete insulin. We generated diabetes-prone non-obese diabetic (NOD) mice that express insulin, via a transgene, in K cells. We assessed the effects on immunogenicity and diabetes development. METHODS: Diabetes incidence and glucose homeostasis were assessed in NOD mice that expressed mouse preproinsulin II from a transgene in K cells and nontransgenic NOD mice (controls); pancreas and duodenum tissues were collected and analyzed by histology. We evaluated T cell responses to insulin, levels of circulating autoantibodies against insulin, and the percentage of circulating antigen-specific T cells. Inflammation of mesenteric and pancreatic lymph node cells was also evaluated. RESULTS: The transgenic mice tended to have lower blood levels of glucose than control mice, associated with increased plasma levels of immunoreactive insulin and proinsulin. Fewer transgenic mice developed diabetes than controls. In analyses of pancreas and intestine tissues from the transgenic mice, insulin-producing K cells were not affected by the immune response and the mice had reduced destruction of endogenous ß cells. Fewer transgenic mice were positive for insulin autoantibodies compared with controls. Cells isolated from mesenteric lymph nodes of the transgenic mice had significantly lower rates of proliferation and T cells from transgenic mice tended to secrete lower levels of inflammatory cytokines than from controls. At 15 weeks, transgenic mice had fewer peripheral CD8(+) T cells specific for NRP-V7 than control mice. CONCLUSIONS: NOD mice with intestinal K cells engineered to express insulin have reduced blood levels of glucose, are less likely to develop diabetes, and have reduced immunity against pancreatic ß cells compared with control NOD mice. This approach might be developed to treat patients with type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/prevenção & controle , Células Enteroendócrinas/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/imunologia , Insulina/metabolismo , Animais , Autoanticorpos/imunologia , Autoanticorpos/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Duodeno/metabolismo , Duodeno/patologia , Células Enteroendócrinas/patologia , Feminino , Homeostase/fisiologia , Insulina/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Pâncreas/metabolismo , Pâncreas/patologia , Linfócitos T/metabolismo , Linfócitos T/patologia
5.
Brain Behav Immun ; 40: 74-84, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24607605

RESUMO

BACKGROUND: Neuropsychiatric disorders during HIV/AIDS are common although the contribution of HIV-1 infection within the brain, and in particular individual HIV-1 proteins, to the development of these brain disorders is unknown. Herein, an in vivo transgenic mouse model was generated in which the HIV-1 Nef protein was expressed in microglia cells, permitting investigation of neurobehavioral phenotypes and associated cellular and molecular properties. METHODS: Transgenic (Tg) mice that expressed full length HIV-1 nef under the control of the c-fms promoter and wildtype (Wt) littermates were investigated using different measures of neurobehavioral performance including locomotory, forced swim (FST), elevated plus maze (EPM) and T-maze tests. Host gene and transgene expression were assessed by RT-PCR, immunoblotting, enzymatic activity and immunohistochemistry. Biogenic amine levels were measured by HPLC with electrochemical detection. RESULTS: Tg animals exhibited Nef expression in brain microglia and cultured macrophages. Tg males displayed hyperactive behaviors including augmented locomotor activity, decreased immobility in the FST and increased open-arm EPM exploration compared to Wt littermates (p<0.05). Tg animals showed increased CCL2 expression with concurrent IFN-α suppression in striatum compared with Wt littermates (p<0.05). Dopamine levels, MAO activity and the dopamine transporter (DAT) expression were reduced in the striatum of Tg animals (p<0.05). CONCLUSIONS: HIV-1 Nef expression in microglia induced CCL2 expression together with disrupting striatal dopaminergic transmission, resulting in hyperactive behaviors which are observed in mania and other psychiatric comorbidities among HIV-infected persons. These findings emphasize the selective effects of individual viral proteins in the brain and their participation in neuropathogenesis.


Assuntos
Dopamina/metabolismo , Microglia/virologia , Atividade Motora , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Animais , Transtorno Bipolar/imunologia , Transtorno Bipolar/virologia , Encéfalo/imunologia , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Feminino , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Microglia/imunologia , Serotonina/metabolismo , Aprendizagem Espacial/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
6.
FASEB J ; 24(6): 1799-812, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20097875

RESUMO

MicroRNAs (miRNAs) are small noncoding RNA molecules, which are known to regulate gene expression in physiological and pathological conditions. miRNA profiling was performed using brain tissue from patients with HIV encephalitis (HIVE), a neuroinflammatory/degenerative disorder caused by HIV infection of the brain. Microarray analysis showed differential expression of multiple miRNAs in HIVE compared to control brains. Target prediction and gene ontology enrichment analysis disclosed targeting of several gene families/biological processes by differentially expressed miRNAs (DEMs), with cell death-related genes, including caspase-6, showing a bias toward down-regulated DEMs. Consistent with the miRNA data, HIVE brains exhibited higher levels of caspase-6 transcripts compared with control patients. Immunohistochemical analysis showed localization of the cleaved form of caspase-6 in astrocytes in HIVE brain sections. Exposure of cultured human primary astrocytes to HIV viral protein R (Vpr) induced p53 up-regulation, loss of mitochondrial membrane potential, and caspase-6 activation followed by cell injury. Transgenic mice, expressing Vpr in microglial cells, demonstrated astrocyte apoptosis in brain, which was associated with caspase-6 activation and neurobehavioral abnormalities. Overall, these data point to previously unrecognized alterations in miRNA profile in the brain during HIV infection, which contribute to cell death through dysregulation of cell death machinery.


Assuntos
Complexo AIDS Demência/etiologia , Astrócitos/citologia , Encéfalo/metabolismo , Caspase 6/metabolismo , Infecções por HIV/complicações , MicroRNAs/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/fisiologia , Adulto , Animais , Astrócitos/metabolismo , Biomarcadores/metabolismo , Western Blotting , Encéfalo/citologia , Sinalização do Cálcio , Caspase 6/genética , Sobrevivência Celular , Feminino , Feto/citologia , Feto/metabolismo , Imunofluorescência , Perfilação da Expressão Gênica , Infecções por HIV/genética , HIV-1 , Humanos , Técnicas Imunoenzimáticas , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Exp Med ; 196(4): 481-92, 2002 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-12186840

RESUMO

Glutamic acid decarboxylase (GAD)65 is an early and important antigen in both human diabetes mellitus and the nonobese diabetic (NOD) mouse. However, the exact role of GAD65-specific T cells in diabetes pathogenesis is unclear. T cell responses to GAD65 occur early in diabetes pathogenesis, yet only one GAD65-specific T cell clone of many identified can transfer diabetes. We have generated transgenic mice on the NOD background expressing a T cell receptor (TCR)-specific for peptide epitope 286-300 (p286) of GAD65. These mice have GAD65-specific CD4(+) T cells, as shown by staining with an I-A(g7)(p286) tetramer reagent. Lymphocytes from these TCR transgenic mice proliferate and make interferon gamma, interleukin (IL)-2, tumor necrosis factor (TNF)-alpha, and IL-10 when stimulated in vitro with GAD65 peptide 286-300, yet these TCR transgenic animals do not spontaneously develop diabetes, and insulitis is virtually undetectable. Furthermore, in vitro activated CD4 T cells from GAD 286 TCR transgenic mice express higher levels of CTL-associated antigen (CTLA)-4 than nontransgenic littermates. CD4(+) T cells, or p286-tetramer(+)CD4(+) Tcells, from GAD65 286-300-specific TCR transgenic mice delay diabetes induced in NOD.scid mice by diabetic NOD spleen cells. This data suggests that GAD65 peptide 286-300-specific T cells have disease protective capacity and are not pathogenic.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus/imunologia , Glutamato Descarboxilase/imunologia , Imunoconjugados , Isoenzimas/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Abatacepte , Animais , Antígenos CD , Antígenos de Diferenciação/genética , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4 , Divisão Celular , Citocinas/biossíntese , Epitopos de Linfócito T/imunologia , Expressão Gênica , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Interleucina-2
8.
J Neurosci ; 27(14): 3703-11, 2007 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-17409234

RESUMO

Despite the introduction of highly active antiretroviral therapy, dementia caused by human immunodeficiency virus-1 (HIV-1) infection remains a devastating and common neurological disorder. Although the mechanisms governing neurodegeneration during HIV-1 infection remain uncertain, the HIV-1 accessory protein, viral protein R (Vpr), has been proposed as a neurotoxic protein. Herein, we report that Vpr protein and transcript were present in the brains of HIV-infected persons. Moreover, soluble Vpr caused neuronal apoptosis, involving cytochrome c extravasation, p53 induction, and activation of caspase-9 while exerting a depressive effect on whole-cell currents in neurons (p < 0.05), which was inhibited by iberiotoxin. Vpr-activated glial cells secreted neurotoxins in a concentration-dependent manner (p < 0.001). Transgenic (Tg) mice expressing Vpr in brain monocytoid cells displayed the transgene principally in the basal ganglia (p < 0.05) and cerebral cortex (p < 0.01) compared with hindbrain expression. Vpr was released from cultured transgenic macrophages, which was cytotoxic to neurons and was blocked by anti-Vpr antibody (p < 0.05). Neuronal injury was observed in Tg animals compared with wild-type littermates, chiefly affecting GAD65 (p < 0.01) and vesicular acetylcholine transferase (p < 0.001) immunopositive neuronal populations in the basal ganglia. There was also a loss of subcortical synaptophysin (p < 0.001) immunoreactivity as well as an increase in activated caspase-3, which was accompanied by a hyperexcitable neurobehavioral phenotype (p < 0.05). Thus, HIV-1 Vpr caused neuronal death through convergent pathogenic mechanisms with ensuing in vivo neurodegeneration, yielding new insights into the mechanisms by which HIV-1 injures the nervous system.


Assuntos
Apoptose/fisiologia , Produtos do Gene vpr/fisiologia , HIV-1/fisiologia , Degeneração Neural/metabolismo , Neurônios/metabolismo , Animais , Linhagem Celular Tumoral , Produtos do Gene vpr/biossíntese , HIV-1/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Degeneração Neural/patologia , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
9.
J Clin Invest ; 115(7): 1879-87, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15937548

RESUMO

The progression of immune responses is generally associated with an increase in the overall avidity of antigen-specific T cell populations for peptide-MHC. This is thought to result from preferential expansion of high-avidity clonotypes at the expense of their low-avidity counterparts. Since T cell antigen-receptor genes do not mutate, it is puzzling that high-avidity clonotypes do not predominate from the outset. Here we provide a developmental basis for this phenomenon in the context of autoimmunity. We have carried out comprehensive studies of the diabetogenic CD8 T cell population that targets residues 206-214 of the beta cell antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP(206-214)) and undergoes avidity maturation as disease progresses. We find that the succession of IGRP(206-214)-specific clonotypes with increasing avidities during the progression of islet inflammation to overt diabetes in nonobese diabetic mice is fueled by autoimmune inflammation but opposed by systemic tolerance. As expected, naive high-avidity IGRP(206-214)-specific T cells respond more efficiently to antigen and are significantly more diabetogenic than their intermediate- or low-avidity counterparts. However, central and peripheral tolerance selectively limit the contribution of these high-avidity T cells to the earliest stages of disease without abrogating their ability to progressively accumulate in inflamed islets and kill beta cells. These results illustrate the way in which incomplete deletion of autoreactive T cell populations of relatively high avidity can contribute to the development of pathogenic autoimmunity in the periphery.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Animais , Autoimunidade , Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Glucose-6-Fosfatase/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Fragmentos de Peptídeos/imunologia , Proteínas/imunologia , Tolerância a Antígenos Próprios
10.
Biochem Biophys Res Commun ; 368(2): 357-63, 2008 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-18222115

RESUMO

The granzyme B gene is activated upon cytotoxic T cell stimulation and the protein is a key inducer of apoptosis in target cells. Previous studies have identified important proximal regulatory regions but these proved insufficient to drive expression in vivo. We identified a DNase1 hypersensitive site (HS2) 3.9kb upstream of the transcription start site that was present in stimulated but not resting CD8+ cells. The CTL line CTLL R8 was stably transfected with GFP reporter constructs and showed consistently higher fluorescence values when HS2 was included. In transgenic mice the presence of the relevant region of DNA resulted in inducible, CTL-specific transcription of the transgene in all transgenic founder lines analyzed. Deletion of HS2 resulted in a 10-fold reduction in expression. This is the first report of a major distal regulatory element in the control of granzyme B transcription.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Regulação Enzimológica da Expressão Gênica/genética , Granzimas/genética , Camundongos Transgênicos/genética , Regiões Promotoras Genéticas/genética , Sequências Reguladoras de Ácido Nucleico/genética , Ativação Transcricional/genética , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
11.
Lymphat Res Biol ; 16(1): 43-55, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29346025

RESUMO

BACKGROUND: Lymphatic endothelial cells (LECs) derived from lymphatic malformations (LMs) bear activated PIK3CA alleles yet display an inflammatory gene expression profile. A basis for the inflammatory phenotype was sought by screening for coexisting somatic mutations. METHODS AND RESULTS: Fourteen independent LEC populations bearing activated PIK3CA alleles were isolated from LM. These were characterized by the expression of growth and inflammatory genes (VEGFC, IL-6, COX-2, IL-8, HO-1, E-SEL) by qRT-PCR. Most commonly upregulated gene products were VEGFC, COX2, HO-1, and ANGPTL4. The specific inhibition of PI3K reduced VEGFC expression without resolving inflammation. Whole exome sequencing of six LM-LEC populations identified five novel somatically acquired alleles coexisting with activated PIK3CA alleles. Two affected genes regulate lipid droplet metabolism (FITM2 and ATG2A), two are gene regulators (MTA1 and TAF1L), and the fifth is an isoform of ANK3 (an endosomal/lysosomal protein). Inhibition of AMPK implicated its involvement in regulating COX-2 and HO-1 overexpression. ANGPTL4 expression was independent of AMPK and PI3K activity and reflected lipid stress demonstrated in normal LECs. AMPK activation with AICAR had a selective growth-limiting effect in a subset of LM-LEC isolates. CONCLUSIONS: Inflammatory stress displayed by LM-LECs is consistent with errors in lipid metabolism that may be linked to acquired mutations. The acquisition of PIK3CA alleles may be a permissive event that antagonizes inflammation and metabolic defect.


Assuntos
Alelos , Classe I de Fosfatidilinositol 3-Quinases/genética , Células Endoteliais/metabolismo , Anormalidades Linfáticas/genética , Tecido Linfoide/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Proteína 4 Semelhante a Angiopoietina/genética , Proteína 4 Semelhante a Angiopoietina/metabolismo , Anquirinas/genética , Anquirinas/metabolismo , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Linhagem Celular , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Selectina E/genética , Selectina E/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Regulação da Expressão Gênica , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Metabolismo dos Lipídeos/genética , Anormalidades Linfáticas/metabolismo , Anormalidades Linfáticas/patologia , Tecido Linfoide/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Cultura Primária de Células , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ribonucleotídeos/farmacologia , Transdução de Sinais , Transativadores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
12.
J Immunol ; 176(12): 7715-25, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16751419

RESUMO

In humans, spontaneous autoimmune attack against cardiomyocytes often leads to idiopathic dilated cardiomyopathy (IDCM) and life-threatening heart failure. HLA-DQ8 transgenic IAb knockout NOD mice (NOD.DQ8/Ab(0); DQA1*0301, DQB1*0302) develop spontaneous anticardiomyocyte autoimmunity with pathology very similar to human IDCM, but why the heart is targeted is unknown. In the present study, we first investigated whether NOD/Ab(0) mice transgenic for a different DQ allele, DQ6, (DQA1*0102, DQB1*0602) would also develop myocarditis. NOD.DQ6/Ab(0) animals showed no cardiac pathology, implying that DQ8 is specifically required for the myocarditis phenotype. To further characterize the cellular immune mechanisms, we established crosses of our NOD.DQ8/Ab(0) animals with Rag1 knockout (Rag1(0)), Ig H chain knockout (IgH(0)), and beta(2)-microglobulin knockout (beta(2)m(0)) lines. Adoptive transfer of purified CD4 T cells from NOD.DQ8/Ab(0) mice with complete heart block (an indication of advanced myocarditis) into younger NOD.DQ8/Ab(0) Rag1(0) animals induced cardiac pathology in all recipients, whereas adoptive transfer of purified CD8 T cells or B lymphocytes had no effect. Despite the absence of B lymphocytes, NOD.DQ8/Ab(0)IgH(0) animals still developed complete heart block, whereas NOD.DQ8/Ab(0)beta(2)m(0) mice (which lack CD8 T cells) failed to develop any cardiac pathology. CD8 T cells (and possibly NK cells) seem to be necessary to initiate disease, whereas once initiated, CD4 T cells alone can orchestrate the cardiac pathology, likely through their capacity to recruit and activate macrophages. Understanding the cellular immune mechanisms causing spontaneous myocarditis/IDCM in this relevant animal model will facilitate the development and testing of new therapies for this devastating disease.


Assuntos
Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/imunologia , Antígenos HLA-DQ/genética , Antígenos de Histocompatibilidade Classe II/genética , Transferência Adotiva , Animais , Doenças Autoimunes/patologia , Linfócitos T CD4-Positivos/transplante , Cardiomiopatia Dilatada/patologia , Modelos Animais de Doenças , Bloqueio Cardíaco/genética , Bloqueio Cardíaco/imunologia , Humanos , Células Matadoras Naturais/imunologia , Transfusão de Linfócitos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Transgênicos
13.
Virology ; 322(1): 69-81, 2004 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-15063118

RESUMO

Clinical and morphologic features of human immunodeficiency virus (HIV)-associated nephropathy (HIVAN), such as proteinuria, sclerosing glomerulopathy, tubular degeneration, and interstitial disease, have been modeled in mice bearing an HIV proviral transgene rendered noninfectious through a deletion in gag/pol. Exploring the genetic basis of HIVAN, HIV transgenic mice bearing mutations in either or both of the accessory genes nef and vpr were created. Proteinuria and focal glomerulosclerosis (FGS) only developed in mice with an intact vpr gene. Transgenic mice bearing a simplified proviral DNA (encoding only Tat and Vpr) developed renal disease characterized by FGS in which Vpr protein was localized to glomerular and tubular epithelia by immunohistochemistry. The dual transgenic progeny of HIV[Tat/Vpr] mice bred to HIV[DeltaVpr] proviral transgenic mice displayed a more severe nephropathy with no apparent increase in Vpr expression, implying that multiple viral genes contribute to HIVAN. However, the unique contribution of macrophage-specific Vpr expression in the development of glomerular disease was underscored by the induction of FGS in multiple murine lines bearing a c-fms/vpr transgene.


Assuntos
Nefropatia Associada a AIDS/virologia , Produtos do Gene vpr/metabolismo , Genes fms , Glomerulosclerose Segmentar e Focal/virologia , HIV-1/genética , Provírus/genética , Nefropatia Associada a AIDS/metabolismo , Animais , DNA Viral/análise , Suscetibilidade a Doenças , Epitélio/metabolismo , Produtos do Gene vpr/biossíntese , Produtos do Gene vpr/genética , Glomerulosclerose Segmentar e Focal/metabolismo , HIV-1/patogenicidade , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Camundongos , Camundongos Transgênicos , Virulência , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA