Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
PLoS Pathog ; 16(10): e1009006, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33057440

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) vGPCR is a constitutively active G protein-coupled receptor that subverts proliferative and inflammatory signaling pathways to induce cell transformation in Kaposi's sarcoma. Cyclooxygenase-2 (COX-2) is an inflammatory mediator that plays a key regulatory role in the activation of tumor angiogenesis. Using two different transformed mouse models and tumorigenic full KSHV genome-bearing cells, including KSHV-Bac16 based mutant system with a vGPCR deletion, we demostrate that vGPCR upregulates COX-2 expression and activity, signaling through selective MAPK cascades. We show that vGPCR expression triggers signaling pathways that upregulate COX-2 levels due to a dual effect upon both its gene promoter region and, in mature mRNA, the 3'UTR region that control mRNA stability. Both events are mediated by signaling through ERK1/2 MAPK pathway. Inhibition of COX-2 in vGPCR-transformed cells impairs vGPCR-driven angiogenesis and treatment with the COX-2-selective inhibitory drug Celecoxib produces a significant decrease in tumor growth, pointing to COX-2 activity as critical for vGPCR oncogenicity in vivo and indicating that COX-2-mediated angiogenesis could play a role in KS tumorigenesis. These results, along with the overexpression of COX-2 in KS lesions, define COX-2 as a potential target for the prevention and treatment of KSHV-oncogenesis.


Assuntos
Herpesvirus Humano 8/metabolismo , Metaloproteinase 2 da Matriz/biossíntese , Receptores Acoplados a Proteínas G/metabolismo , Sarcoma de Kaposi/irrigação sanguínea , Animais , Carcinogênese , Transformação Celular Neoplásica/genética , Células Endoteliais/metabolismo , Proteínas de Ligação ao GTP/genética , Herpesvirus Humano 8/genética , Sistema de Sinalização das MAP Quinases , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Nus , Células NIH 3T3 , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/virologia , Oncogenes , Receptores Acoplados a Proteínas G/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Transdução de Sinais , Ativação Transcricional
2.
Bio Protoc ; 9(8)2019 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-32864391

RESUMO

Kaposi's sarcoma (KS) herpesvirus (KSHV) is a virus that causes KS, an angiogenic AIDS-associated spindle-cell neoplasm, by activating host oncogenic signaling cascades through autocrine and paracrine mechanisms. Many host signaling cascades co-opted by KSHV including PI3K/AKT/mTORC, NFkB and Notch are critical for cell-specific mechanisms of transformation and their identification is paving the way to therapeutic target discovery. Analysis of the molecular KS signature common to human KS tumors and our mouse KS-like tumors showed consistent expression of KS markers VEGF and PDGF receptors with upregulation of other angiogenesis ligands and their receptors in vivo. This points to the autocrine and paracrine activation of various receptor tyrosine kinase (RTK) signaling axes. Hereby we describe a protocol to screen for activated receptor tyrosine kinase of KSHV-induced KS-like mouse tumors using a Mouse Phospho-RTK Array Kit and its validation by RTK western blots. We showed that this method can be successfully used to rank the tyrosine kinase receptors most activated in tumors in an unbiased manner. This allowed us to identify PDGFRA as an oncogenic driver and therapeutic target in AIDS-KS.

3.
Rev. bras. entomol ; 62(4): 275-282, Oct.-Dec. 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1045532

RESUMO

ABSTRACT The replacement of natural grassland by cultivated areas might favor the increase in abundance of some root-feeding species such as the white grubs, which may become a constraint for field crop production. This research aimed to assay the population density and geographical distribution of white grubs pest and other species in natural grassland and cultivated areas throughout the Brazilian Pampa biome. White grubs were sampled in 18 locations in both landscape use types and identified. Population density (number of larvae m-2) was calculated for each recorded species and sorted within two groups (pest species and other species), compared between natural grasslands and cultivated areas, as well as among locations. A dendrogram to evaluate species similarity among locations was built based on combined data obtained from both landscape use types throughout the region. In total, 31 species were found in the Brazilian Pampa, and four of them are considered as crop pests: Diloboderus abderus (Sturm, 1826), Euetheola humilis (Burmeister, 1847), Lyogenys fusca (Blanchard, 1830), and Phyllophaga triticophaga Morón & Salvadori, 1998. The average population density of pest species in cultivated areas was less than five larvae m-2, at most of locations. Some species had a wide geographical distribution (e.g. D. abderus and Cyclocephala modesta Burmeister), while other melolontids occurred at only one location. The knowledge of which white grub species are present in a field and its population densities assist farmers to take proper management decisions.

4.
PLoS Pathog ; 14(7): e1007175, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29985958

RESUMO

Kaposi's sarcoma (KS) herpesvirus (KSHV) causes KS, an angiogenic AIDS-associated spindle-cell neoplasm, by activating host oncogenic signaling cascades through autocrine and paracrine mechanisms. Tyrosine kinase receptor (RTK) proteomic arrays, identified PDGF receptor-alpha (PDGFRA) as the predominantly-activated RTK in KSHV-induced mouse KS-tumors. We show that: 1) KSHV lytic replication and the vGPCR can activate PDGFRA through upregulation of its ligands PDGFA/B, which increase c-myc, VEGF and KSHV gene expression in infected cells 2) KSHV infected spindle cells of most AIDS-KS lesions display robust phospho-PDGFRA staining 3) blocking PDGFRA-signaling with N-acetyl-cysteine, RTK-inhibitors Imatinib and Sunitinib, or dominant-negative PDGFRA inhibits tumorigenesis 4) PDGFRA D842V activating-mutation confers resistance to Imatinib in mouse-KS tumorigenesis. Our data show that KSHV usurps sarcomagenic PDGFRA signaling to drive KS. This and the fact that PDGFRA drives non-viral sarcomas highlights the importance for KSHV-induced ligand-mediated activation of PDGFRA in KS sarcomagenesis and shows that this oncogenic axis could be successfully blocked to impede KS tumor growth.


Assuntos
Carcinogênese/metabolismo , Herpesvirus Humano 8/patogenicidade , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Sarcoma de Kaposi/virologia , Animais , Humanos , Camundongos , Camundongos Nus , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Sarcoma de Kaposi/metabolismo , Transdução de Sinais
7.
Immunol Res ; 57(1-3): 159-65, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24203444

RESUMO

Kaposi's sarcoma herpesvirus or human herpesvirus-8 (KSHV/HHV-8) is the etiological agent of Kaposi's sarcoma (KS), an AIDS-defining angioproliferative neoplasm that continues to be a major global health problem and, of primary effusion lymphoma (PEL), a rare incurable B-cell lymphoma. This review describes the research from our laboratory and its collaborators to uncover molecular mechanisms of viral oncogenesis in order to develop new pathogenesis-based therapies to the KSHV-induced AIDS malignancies KS and PEL. They include the discovery of the viral angiogenic oncogene G protein-coupled receptor (vGPCR), the development of mouse models of KSHV and oxidative stress-induced KS, the identification of the role of Rac1-induced ROS in viral oncogenesis of KS and the development of novel therapeutic approaches able to target both latent and lytic oncogenic KSHV infection.


Assuntos
Transformação Celular Viral , Infecções por Herpesviridae/complicações , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/etiologia , Animais , Modelos Animais de Doenças , Humanos , Estresse Oxidativo , Comunicação Parácrina , Receptores Acoplados a Proteínas G/metabolismo , Sarcoma de Kaposi/metabolismo , Pesquisa Translacional Biomédica , Proteínas rac1 de Ligação ao GTP/metabolismo
8.
Antioxid Redox Signal ; 18(1): 80-90, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22746102

RESUMO

AIMS: Kaposi's sarcoma (KS), caused by the Kaposi's sarcoma herpesvirus (KSHV), is an AIDS-associated cancer characterized by angiogenesis and proliferation of spindle cells. Rac1-activated reactive oxygen species (ROS) production has been implicated in KS tumorigenesis. We used an animal model of KSHV-induced Kaposi's sarcomagenesis (mECK36) to study the role of ROS in KS and the efficacy of N-acetyl l-cysteine (NAC) in inhibiting or preventing KS. RESULTS: Signaling by the KSHV early lytic gene viral G protein-coupled receptor (vGPCR) activated ROS production in mECK36 cells via a Rac1-NADPH oxidase pathway. Induction of the lytic cycle in KSHV-infected KS spindle cells upregulated ROS along with upregulation of vGPCR expression. We also found that expression of the major latent transcript in 293 cells increased ROS levels. ROS scavenging with NAC halted mECK36 tumor growth in a KSHV-specific manner. NAC inhibited KSHV latent gene expression as well as tumor angiogenesis and lymphangiogenesis. These effects correlated with the reduction of vascular endothelial growth factor (VEGF), c-myc, and cyclin D1, and could be explained on the basis of inhibition of STAT3 tyrosine phosphorylation. NAC prevented mECK36 de novo tumor formation. Molecular analysis of NAC-resistant tumors revealed a strong upregulation of Rac1 and p40(PHOX). INNOVATION AND CONCLUSION: Our results demonstrate that ROS-induction by KSHV plays a causal role in KS oncogenesis by promoting proliferation and angiogenesis. Our results show that both ROS and their molecular sources can be targeted therapeutically using NAC or other Food and Drug Administration (FDA)-approved inhibitors for prevention and treatment of AIDS-KS.


Assuntos
Transformação Celular Neoplásica/metabolismo , Herpesvirus Humano 8/genética , Espécies Reativas de Oxigênio/metabolismo , Sarcoma de Kaposi/virologia , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Inibidores da Angiogênese/farmacologia , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Proliferação de Células , Transformação Celular Neoplásica/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Herpesvirus Humano 8/efeitos dos fármacos , Herpesvirus Humano 8/fisiologia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica/prevenção & controle , Neuropeptídeos/metabolismo , Estresse Oxidativo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/prevenção & controle , Transcrição Gênica/efeitos dos fármacos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP
9.
J Exp Med ; 209(11): 1985-2000, 2012 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-23027923

RESUMO

Kaposi's sarcoma (KS), a multifocal vascular neoplasm linked to human herpesvirus-8 (HHV-8/KS-associated herpesvirus [KSHV]) infection, is the most common AIDS-associated malignancy. Clinical management of KS has proven to be challenging because of its prevalence in immunosuppressed patients and its unique vascular and inflammatory nature that is sustained by viral and host-derived paracrine-acting factors primarily released under hypoxic conditions. We show that interactions between the regulatory lectin galectin-1 (Gal-1) and specific target N-glycans link tumor hypoxia to neovascularization as part of the pathogenesis of KS. Expression of Gal-1 is found to be a hallmark of human KS but not other vascular pathologies and is directly induced by both KSHV and hypoxia. Interestingly, hypoxia induced Gal-1 through mechanisms that are independent of hypoxia-inducible factor (HIF) 1α and HIF-2α but involved reactive oxygen species-dependent activation of the transcription factor nuclear factor κB. Targeted disruption of Gal-1-N-glycan interactions eliminated hypoxia-driven angiogenesis and suppressed tumorigenesis in vivo. Therapeutic administration of a Gal-1-specific neutralizing mAb attenuated abnormal angiogenesis and promoted tumor regression in mice bearing established KS tumors. Given the active search for HIF-independent mechanisms that serve to couple tumor hypoxia to pathological angiogenesis, our findings provide novel opportunities not only for treating KS patients but also for understanding and managing a variety of solid tumors.


Assuntos
Galectina 1/metabolismo , Neovascularização Patológica/metabolismo , Polissacarídeos/metabolismo , Sarcoma de Kaposi/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Hipóxia Celular , Linhagem Celular Tumoral , Células Cultivadas , Galectina 1/genética , Galectina 1/imunologia , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Hipóxia , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Patológica/prevenção & controle , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma de Kaposi/tratamento farmacológico , Sarcoma de Kaposi/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Proc Natl Acad Sci U S A ; 107(29): 13069-74, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20615981

RESUMO

Primary effusion lymphoma (PEL) is an aggressive B-cell lymphoma most commonly diagnosed in HIV-positive patients and universally associated with Kaposi's sarcoma-associated herpesvirus (KSHV). Chemotherapy treatment of PEL yields only short-term remissions in the vast majority of patients, but efforts to develop superior therapeutic approaches have been impeded by lack of animal models that accurately mimic human disease. To address this issue, we developed a direct xenograft model, UM-PEL-1, by transferring freshly isolated human PEL cells into the peritoneal cavities of NOD/SCID mice without in vitro cell growth to avoid the changes in KSHV gene expression evident in cultured cells. We used this model to show that bortezomib induces PEL remission and extends overall survival of mice bearing lymphomatous effusions. The proapoptotic effects of bortezomib are not mediated by inhibition of the prosurvival NF-kappaB pathway or by induction of a terminal unfolded protein response. Transcriptome analysis by genomic arrays revealed that bortezomib down-regulated cell-cycle progression, DNA replication, and Myc-target genes. Furthermore, we demonstrate that in vivo treatment with either bortezomib or doxorubicin induces KSHV lytic reactivation. These reactivations were temporally distinct, and this difference may help elucidate the therapeutic window for use of antivirals concurrently with chemotherapy. Our findings show that this direct xenograft model can be used for testing novel PEL therapeutic strategies and also can provide a rational basis for evaluation of bortezomib in clinical trials.


Assuntos
Ácidos Borônicos/uso terapêutico , Linfoma de Efusão Primária/tratamento farmacológico , Pirazinas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Idoso de 80 Anos ou mais , Animais , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Bortezomib , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Chlorocebus aethiops , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/genética , Regulação para Baixo/efeitos dos fármacos , Fator de Transcrição E2F3/metabolismo , Evolução Fatal , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 8/efeitos dos fármacos , Herpesvirus Humano 8/fisiologia , Humanos , Linfoma de Efusão Primária/virologia , Masculino , Camundongos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pirazinas/farmacologia , Análise de Sobrevida , Resultado do Tratamento , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Células Vero , Vírion/efeitos dos fármacos , Vírion/metabolismo
11.
Cell Host Microbe ; 6(3): 218-30, 2009 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-19748464

RESUMO

The HIV Nef protein mediates endocytosis of surface receptors that correlates with disease progression, but the link between this Nef function and HIV pathogenesis is not clear. Here, we report that Nef-mediated activation of membrane trafficking is bidirectional, connecting endocytosis with exocytosis as occurs in activated T cells. Nef expression induced an extensive secretory activity in infected and, surprisingly, also in noninfected T cells, leading to the massive release of microvesicle clusters, a phenotype observed in vitro and in 36%-87% of primary CD4 T cells from HIV-infected individuals. Consistent with exocytosis in noninfected cells, Nef is transferred to bystander cells upon cell-to-cell contact and subsequently induces secretion in an Erk1/2-dependent manner. Thus, HIV Nef alters membrane dynamics, mimicking those of activated T cells and causing a transfer of infected cell signaling (TOS) to bystander cells. This mechanism may help explain the detrimental effect on bystander cells seen in HIV infection.


Assuntos
Efeito Espectador , Exocitose , Infecções por HIV/metabolismo , HIV-1/metabolismo , Linfócitos T/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Células Cultivadas , Endocitose , Infecções por HIV/imunologia , Interações Hospedeiro-Patógeno , Humanos , Ativação Linfocitária , Transporte Proteico , Via Secretória , Vesículas Secretórias/metabolismo
12.
Proc Natl Acad Sci U S A ; 106(21): 8683-8, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19429708

RESUMO

Kaposi's sarcoma (KS) is the major AIDS-associated malignancy. It is characterized by the proliferation of spindle cells, inflammatory infiltrate, and aberrant angiogenesis caused by Kaposi's sarcoma herpesvirus (KSHV) infection. Small GTPase Rac1, an inflammatory signaling mediator triggering reactive oxygen species (ROS) production by NADPH-oxidases, is implicated in carcinogenesis and tumor angiogenesis. Here, we show that expression of a constitutively active Rac1 (RacCA) driven by the alpha-smooth muscle actin promoter in transgenic mice is sufficient to cause KS-like tumors through mechanisms involving ROS-driven proliferation, up-regulation of AKT signaling, and hypoxia-inducible factor 1-alpha-related angiogenesis. RacCA-induced tumors expressed KS phenotypic markers; displayed remarkable transcriptome overlap with KS lesions; and were, like KS, associated with male gender. The ROS scavenging agent N-acetyl-cysteine inhibited angiogenesis and completely abrogated transgenic RacCA tumor formation, indicating a causal role of ROS in tumorigenesis. Consistent with a pathogenic role in KS, immunohistochemical analysis revealed that Rac1 is overexpressed in KSHV(+) spindle cells of AIDS-KS biopsies. Our results demonstrate the direct oncogenicity of Rac1 and ROS and their contribution to a KS-like malignant phenotype, further underscoring the carcinogenic potential of oxidative stress in the context of chronic infection and inflammation. They define the RacCA transgenic mouse as a model suitable for studying the role of oxidative stress in the pathogenesis and therapy of KS, with relevance to other inflammation-related malignancies. Our findings suggest host and viral genes triggering Rac1 or ROS production as key determinants of KS onset and potential KS chemopreventive or therapeutic targets.


Assuntos
Antioxidantes/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Síndrome da Imunodeficiência Adquirida/complicações , Síndrome da Imunodeficiência Adquirida/enzimologia , Síndrome da Imunodeficiência Adquirida/genética , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Ativação Enzimática , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sarcoma de Kaposi/irrigação sanguínea , Sarcoma de Kaposi/etiologia , Transcrição Gênica/genética , Células Tumorais Cultivadas , Proteínas rac1 de Ligação ao GTP/genética
13.
Biosci Rep ; 29(1): 25-34, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18680481

RESUMO

Giardia lamblia is a medically important protozoan parasite with a basal position in the eukaryotic lineage and is an interesting model to explain the evolution of biochemical events in eukaryotic cells. G. lamblia trophozoites undergo significant changes in order to survive outside the intestine of their host by differentiating into infective cysts. In the present study, we characterize the previously identified Orf-C4 (G. lamblia open reading frame C4) gene, which is considered to be specific to G. lamblia. It encodes a 22 kDa protein that assembles into high-molecular-mass complexes during the entire life cycle of the parasite. ORF-C4 localizes to the cytoplasm of trophozoites and cysts, and forms large spherical aggregates when overexpressed. ORF-C4 overexpression and down-regulation do not affect trophozoite viability; however, differentiation into cysts is slightly delayed when the expression of ORF-C4 is down-regulated. In addition, ORF-C4 protein expression is modified under specific stress-inducing conditions. Neither orthologous proteins nor conserved domains are found in databases by conventional sequence analysis of the predicted protein. However, ORF-C4 contains a region which is similar structurally to the alpha-crystallin domain of sHsps (small heat-shock proteins). In the present study, we show the potential role of ORF-C4 as a small chaperone which is involved in the response to stress (including encystation) in G. lamblia.


Assuntos
Giardia lamblia/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Animais , Regulação da Expressão Gênica , Giardia lamblia/genética , Proteínas de Choque Térmico Pequenas/genética , Proteínas de Choque Térmico Pequenas/metabolismo , Dados de Sequência Molecular , Análise de Sequência de DNA , Estresse Fisiológico , alfa-Cristalinas/genética , alfa-Cristalinas/metabolismo
14.
Cancer Cell ; 11(3): 245-58, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17349582

RESUMO

Transfection of a Kaposi's sarcoma (KS) herpesvirus (KSHV) Bacterial Artificial Chromosome (KSHVBac36) into mouse bone marrow endothelial-lineage cells generates a cell (mECK36) that forms KS-like tumors in mice. mECK36 expressed most KSHV genes and were angiogenic, but they didn't form colonies in soft agar. In nude mice, mECK36 formed KSHV-harboring vascularized spindle cell sarcomas that were LANA+/podoplanin+, overexpressed VEGF and Angiopoietin ligands and receptors, and displayed KSHV and host transcriptomes reminiscent of KS. mECK36 that lost the KSHV episome reverted to nontumorigenicity. siRNA suppression of KSHV vGPCR, an angiogenic gene upregulated in mECK36 tumors, inhibited angiogenicity and tumorigenicity. These results show that KSHV malignancy is in vivo growth restricted and reversible, defining mECK36 as a biologically sensitive animal model of KSHV-dependent KS.


Assuntos
Modelos Animais de Doenças , Herpesvirus Humano 8 , Sarcoma de Kaposi/patologia , Angiopoietinas/metabolismo , Animais , Antígenos Virais/metabolismo , Células da Medula Óssea/patologia , Linhagem da Célula , Transformação Celular Neoplásica , Transformação Celular Viral , Células Cultivadas , Cromossomos Artificiais Bacterianos , Células Endoteliais/patologia , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica , Proteínas Nucleares/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...