Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
2.
Mol Ther Methods Clin Dev ; 31: 101135, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38027064

RESUMO

Immunotherapy of acute myeloid leukemia (AML) has been challenging because the lack of tumor-specific antigens results in "on-target, off-tumor" toxicity. To unlock the full potential of AML therapies, we used CRISPR-Cas9 to genetically ablate the myeloid protein CD33 from healthy donor hematopoietic stem and progenitor cells (HSPCs), creating tremtelectogene empogeditemcel (trem-cel). Trem-cel is a HSPC transplant product designed to provide a reconstituted hematopoietic compartment that is resistant to anti-CD33 drug cytotoxicity. Here, we describe preclinical studies and process development of clinical-scale manufacturing of trem-cel. Preclinical data showed proof-of-concept with loss of CD33 surface protein and no impact on myeloid cell differentiation or function. At clinical scale, trem-cel could be manufactured reproducibly, routinely achieving >70% CD33 editing with no effect on cell viability, differentiation, and function. Trem-cel pharmacology studies using mouse xenograft models showed long-term engraftment, multilineage differentiation, and persistence of gene editing. Toxicology assessment revealed no adverse findings, and no significant or reproducible off-target editing events. Importantly, CD33-knockout myeloid cells were resistant to the CD33-targeted agent gemtuzumab ozogamicin in vitro and in vivo. These studies supported the initiation of the first-in-human, multicenter clinical trial evaluating the safety and efficacy of trem-cel in patients with AML (NCT04849910).

3.
Elife ; 42015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25714926

RESUMO

Angiopoietin-like proteins (angptls) are capable of ex vivo expansion of mouse and human hematopoietic stem and progenitor cells (HSPCs). Despite this intriguing ability, their mechanism is unknown. In this study, we show that angptl2 overexpression is sufficient to expand definitive HSPCs in zebrafish embryos. Angptl1/2 are required for definitive hematopoiesis and vascular specification of the hemogenic endothelium. The loss-of-function phenotype is reminiscent of the notch mutant mindbomb (mib), and a strong genetic interaction occurs between angptls and notch. Overexpressing angptl2 rescues mib while overexpressing notch rescues angptl1/2 morphants. Gene expression studies in ANGPTL2-stimulated CD34(+) cells showed a strong MYC activation signature and myc overexpression in angptl1/2 morphants or mib restored HSPCs formation. ANGPTL2 can increase NOTCH activation in cultured cells and ANGPTL receptor interacted with NOTCH to regulate NOTCH cleavage. Together our data provide insight to the angptl-mediated notch activation through receptor interaction and subsequent activation of myc targets.


Assuntos
Angiopoietinas/genética , Células-Tronco Hematopoéticas/metabolismo , Receptores Notch/genética , Transdução de Sinais/genética , Proteínas de Peixe-Zebra/genética , Proteína 1 Semelhante a Angiopoietina , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/metabolismo , Animais , Animais Geneticamente Modificados , Western Blotting , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Células HEK293 , Hematopoese/genética , Humanos , Células K562 , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Microscopia Confocal , Ligação Proteica , Interferência de RNA , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Imagem com Lapso de Tempo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
4.
J Cell Sci ; 126(Pt 24): 5541-52, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24046447

RESUMO

Transient disruption of endothelial adherens junctions and cytoskeletal remodeling are responsible for increases in vascular permeability induced by inflammatory stimuli and vascular endothelial growth factor (VEGF). Nitric oxide (NO) produced by endothelial NO synthase (eNOS) is crucial for VEGF-induced changes in permeability in vivo; however, the molecular mechanism by which endogenous NO modulates endothelial permeability is not clear. Here, we show that the lack of eNOS reduces VEGF-induced permeability, an effect mediated by enhanced activation of the Rac GTPase and stabilization of cortical actin. The loss of NO increased the recruitment of the Rac guanine-nucleotide-exchange factor (GEF) TIAM1 to adherens junctions and VE-cadherin (also known as cadherin 5), and reduced Rho activation and stress fiber formation. In addition, NO deficiency reduced VEGF-induced VE-cadherin phosphorylation and impaired the localization, but not the activation, of c-Src to cell junctions. The physiological role of eNOS activation is clear given that VEGF-, histamine- and inflammation-induced vascular permeability is reduced in mice bearing a non-phosphorylatable knock-in mutation of the key eNOS phosphorylation site S1176. Thus, NO is crucial for Rho GTPase-dependent regulation of cytoskeletal architecture leading to reversible changes in vascular permeability.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Endotélio Vascular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/fisiologia , Junções Aderentes/metabolismo , Animais , Proteína Tirosina Quinase CSK , Permeabilidade Capilar , Células Cultivadas , Células Endoteliais/enzimologia , Endotélio Vascular/citologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/genética , Fosforilação , Processamento de Proteína Pós-Traducional , Transporte Proteico , Fibras de Estresse/metabolismo , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Fator A de Crescimento do Endotélio Vascular/fisiologia , Quinases da Família src/metabolismo
5.
Am J Pathol ; 182(5): 1900-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23499551

RESUMO

Cancer cells are often characterized by high proliferation rates, a consequence of increased mitotic signaling coupled with unchecked cellular growth. We recently demonstrated that vascular endothelial cells unexpectedly express ferlins, a family of muscle-specific proteins capable of regulating the fusion of lipid patches to the plasma membrane, and that these highly regulated membrane fusion events are essential to endothelial cell proliferation and homeostasis. Here, we show that human and mouse breast cancer cell lines also express myoferlin at various levels, and that the processes of transformation, epithelial-mesenchymal transition, and metastasis do not appear to have any effect on myoferlin expression in vitro. In vivo, we observed that solid mouse and human carcinoma tissues also express high levels of myoferlin protein. Loss-of-function studies performed in mice revealed that myoferlin gene knockdown can attenuate cancer cell proliferation in vitro and decrease tumor burden, and that accelerated tumor cell growth appears to rely on intact myoferlin-dependent membrane repair and signaling under exponential growth conditions. To our knowledge, these data provide the first evidence of myoferlin expression in solid human and mouse tumors. We have thus identified a novel membrane repair process that likely helps sustain the high growth rates characteristic of tumors, and we suggest that interfering with normal myoferlin expression and/or membrane repair and remodeling may provide therapeutically relevant antiproliferative effects.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Carcinogênese/metabolismo , Carcinogênese/patologia , Carcinoma/metabolismo , Carcinoma/patologia , Membrana Celular/patologia , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Idoso , Animais , Carcinoma/irrigação sanguínea , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Feminino , Técnicas de Silenciamento de Genes , Humanos , Camundongos
6.
Proc Natl Acad Sci U S A ; 110(2): E141-50, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23236128

RESUMO

During development, the hematopoietic lineage transits through hemogenic endothelium, but the signaling pathways effecting this transition are incompletely characterized. Although the Hedgehog (Hh) pathway is hypothesized to play a role in patterning blood formation, early embryonic lethality of mice lacking Hh signaling precludes such analysis. To determine a role for Hh signaling in patterning of hemogenic endothelium, we assessed the effect of altered Hh signaling in differentiating mouse ES cells, cultured mouse embryos, and developing zebrafish embryos. In differentiating mouse ES cells and mouse yolk sac cultures, addition of Indian Hh ligand increased hematopoietic progenitors, whereas chemical inhibition of Hh signaling reduced hematopoietic progenitors without affecting primitive streak mesoderm formation. In the setting of Hh inhibition, induction of either Notch signaling or overexpression of Stem cell leukemia (Scl)/T-cell acute lymphocytic leukemia protein 1 rescued hemogenic vascular-endothelial cadherin(+) cells and hematopoietic progenitor formation. Together, our results reveal that Scl overexpression is sufficient to rescue the developmental defects caused by blocking the Hh and Notch pathways, and inform our understanding of the embryonic endothelial-to-hematopoietic transition.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Endotélio/fisiologia , Proteínas Hedgehog/metabolismo , Células-Tronco Hematopoéticas/citologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Ensaio de Unidades Formadoras de Colônias , Embrião de Mamíferos , Feminino , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/fisiologia , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Peixe-Zebra
7.
Blood ; 120(16): 3371-81, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22936663

RESUMO

Angiogenic sprouting requires that cell-cell contacts be maintained during migration of endothelial cells. Angiopoietin-1 (Ang-1) and vascular endothelial growth factor act oppositely on endothelial cell junctions. We found that Ang-1 promotes collective and directional migration and, in contrast to VEGF, induces the formation of a complex formed of atypical protein kinase C (PKC)-ζ and ß-catenin at cell-cell junctions and at the leading edge of migrating endothelial cells. This complex brings Par3, Par6, and adherens junction proteins at the front of migrating cells to locally activate Rac1 in response to Ang-1. The colocalization of PKCζ and ß-catenin at leading edge along with PKCζ-dependent stabilization of cell-cell contacts promotes directed and collective endothelial cell migration. Consistent with these results, down-regulation of PKCζ in endothelial cells alters Ang-1-induced sprouting in vitro and knockdown in developing zebrafish results in intersegmental vessel defects caused by a perturbed directionality of tip cells and by loss of cell contacts between tip and stalk cells. These results reveal that PKCζ and ß-catenin function in a complex at adherens junctions and at the leading edge of migrating endothelial cells to modulate collective and directional migration during angiogenesis.


Assuntos
Angiopoietina-1/farmacologia , Movimento Celular/fisiologia , Endotélio Vascular/metabolismo , Neovascularização Fisiológica/fisiologia , Proteína Quinase C/metabolismo , beta Catenina/metabolismo , Junções Aderentes/metabolismo , Animais , Animais Geneticamente Modificados , Aorta/citologia , Aorta/metabolismo , Células COS , Bovinos , Movimento Celular/efeitos dos fármacos , Polaridade Celular , Células Cultivadas , Chlorocebus aethiops , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Endotélio Vascular/citologia , Imunofluorescência , Junções Intercelulares/metabolismo , Microinjeções , Cicatrização , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
8.
J Pediatr Endocrinol Metab ; 24(9-10): 771-3, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22145473

RESUMO

Pituitary abscess is a rare but potentially life-threatening infectious process. Diagnosis is challenging as symptoms are non-specific and signs of infection may be absent. We report the case of a previously healthy 17-year-old male who presented with worsening headaches, polyuria, polydipsia and no clinical signs of infection. On evaluation, he was found to have hypopituitarism with diabetes insipidus, hypothyroidism and adrenal insufficiency. An imaging study revealed a pituitary mass. He underwent transsphenoidal biopsy to rule out tumor. The abscess was drained transsphenoidally and he was treated with parental antibiotics. Magnetic resonance imaging one year later revealed a normal pituitary without any evidence of abscess or mass. He continues to require thyroid, adrenal and anti-diuretic hormone replacements. As with any pituitary lesion, prompt complete hypothalamic pituitary evaluation is essential to avoid potentially life-threatening consequences.


Assuntos
Abscesso Encefálico/patologia , Diabetes Insípido/patologia , Hipopituitarismo/patologia , Imageamento por Ressonância Magnética , Doenças da Hipófise/patologia , Adolescente , Abscesso Encefálico/complicações , Abscesso Encefálico/terapia , Diabetes Insípido/etiologia , Drenagem , Humanos , Hipopituitarismo/etiologia , Masculino , Doenças da Hipófise/etiologia
9.
Haematologica ; 96(12): 1753-60, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21933862

RESUMO

BACKGROUND: Controversy persists regarding the role of Notch signaling in myelopoiesis. We have used genetic approaches, employing two Notch zebrafish mutants deadly seven (DES) and beamter (BEA) with disrupted function of notch1a and deltaC, respectively, and Notch1a morphants to analyze the development of leukocyte populations in embryonic and mature fish. DESIGN AND METHODS: Myelomonocytes were quantified in early embryos by in situ hybridization using a myeloper-oxidase (mpx) probe. Morpholinos were used to knock down expression of Notch1a or DeltaC. Wound healing assays and/or flow cytometry were used to quantify myelomonocytes in 5-day post-fertilization (dpf) Notch mutants (BEA and DES), morphants or pu.1:GFP, mpx:GFP and fms:RFP transgenic embryos. Flow cytometry was performed on 2-3 month old mutant fish. RESULTS: The number of mpx(+) cells in embryos was reduced at 48 hpf (but not at 26 hpf) in DES compared to WT. At 5 dpf this was reflected by a reduction in the number of myelomonocytic cells found at the wound site in mutants and in Notch1a morphants. This was due to a reduced number of myelomonocytes developing rather than a deficit in the migratory ability since transient inhibition of Notch signaling using DAPT had no effect. The early deficit in myelopoiesis was maintained into later life, 2-3 month old BEA and DES fish having a decreased proportion of myelomonocytes in both the hematopoietic organ (kidney marrow) and the periphery (coelomic cavity). CONCLUSIONS: Our results indicate that defects in Notch signaling affect definitive hematopoiesis, altering myelopoiesis from the early stages of development into the adult.


Assuntos
Embrião não Mamífero/embriologia , Proteínas de Homeodomínio/metabolismo , Mielopoese/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Embrião não Mamífero/citologia , Proteínas de Homeodomínio/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Organismos Geneticamente Modificados/embriologia , Organismos Geneticamente Modificados/genética , Receptor Notch1/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
10.
Mol Cell Proteomics ; 9(10): 2109-24, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20585024

RESUMO

Caveolae are organelles abundant in the plasma membrane of many specialized cells including endothelial cells (ECs), epithelial cells, and adipocytes, and in these cells, caveolin-1 (Cav-1) is the major coat protein essential for the formation of caveolae. To identify proteins that require Cav-1 for stable incorporation into membrane raft domains, a quantitative proteomics analysis using isobaric tagging for relative and absolute quantification was performed on rafts isolated from wild-type and Cav-1-deficient mice. In three independent experiments, 117 proteins were consistently identified in membrane rafts with the largest differences in the levels of Cav-2 and in the caveola regulatory proteins Cavin-1 and Cavin-2. Because the lung is highly enriched in ECs, we validated and characterized the role of the newly described protein Cavin-1 in several cardiovascular tissues and in ECs. Cavin-1 was highly expressed in ECs lining blood vessels and in cultured ECs. Knockdown of Cavin-1 reduced the levels of Cav-1 and -2 and weakly influenced the formation of high molecular weight oligomers containing Cav-1 and -2. Cavin-1 silencing enhanced basal nitric oxide release from ECs but blocked proangiogenic phenotypes such as EC proliferation, migration, and morphogenesis in vitro. Thus, these data support an important role of Cavin-1 as a regulator of caveola function in ECs.


Assuntos
Caveolina 1/metabolismo , DNA Polimerase I/metabolismo , Proteômica , Animais , Sequência de Bases , Western Blotting , Caveolina 1/genética , Linhagem Celular , Movimento Celular , Proliferação de Células , Cromatografia por Troca Iônica , Inativação Gênica , Humanos , Espectrometria de Massas , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Óxido Nítrico/metabolismo , RNA Interferente Pequeno
11.
Proc Natl Acad Sci U S A ; 105(50): 20009-14, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19060214

RESUMO

Although COX-dependent production of prostaglandins (PGs) is known to be crucial for tumor angiogenesis and growth, the role of PGD(2) remains virtually unknown. Here we show that PGD(2) receptor (DP) deficiency enhances tumor progression accompanied by abnormal vascular expansion. In tumors, angiogenic endothelial cells highly express DP receptor, and its deficiency accelerates vascular leakage and angiogenesis. Administration of a synthetic DP agonist, BW245C, markedly suppresses tumor growth as well as tumor hyperpermeability in WT mice, but not in DP-deficient mice. In a corneal angiogenesis assay and a modified Miles assay, host DP deficiency potentiates angiogenesis and vascular hyperpermeability under COX-2-active situation, whereas exogenous administration of BW245C strongly inhibits both angiogenic properties in WT mice. In an in vitro assay, BW245C does not affect endothelial migration and tube formation, processes that are necessary for angiogenesis; however, it strongly improves endothelial barrier function via an increase in intracellular cAMP production. Our results identify PGD(2)/DP receptor as a new regulator of tumor vascular permeability, indicating DP agonism may be exploited as a potential therapy for the treatment of cancer.


Assuntos
Permeabilidade Capilar , Endotélio Vascular/metabolismo , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Animais , Permeabilidade Capilar/efeitos dos fármacos , Movimento Celular , Proliferação de Células , Neovascularização da Córnea/induzido quimicamente , AMP Cíclico/metabolismo , Endotélio Vascular/patologia , Hidantoínas/farmacologia , Interleucina-1beta/farmacologia , Camundongos , Camundongos Mutantes , Receptores Imunológicos/agonistas , Receptores Imunológicos/genética , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia
12.
Arterioscler Thromb Vasc Biol ; 28(1): 105-11, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17975117

RESUMO

OBJECTIVE: Heat-shock protein 90 (Hsp90) coordinates the regulation of diverse signaling proteins. We try to develop a new tool to explore the regulatory functions of Hsp90 in endothelial cells (ECs) instead of the existing chemical approaches. METHODS AND RESULTS: We designed a dominant-negative Hsp90 construct by site-direct mutagenesis of residue Asp-88 to Asn (D88N-Hsp90) based on the structure of the ATP/ADP-binding site. Recombinant wild-type Hsp90 protein binds ATP-Sepharose beads in manner inhibited by ATP or 17-AAG, a specific inhibitor for Hsp90, however the binding activity of D88N-Hsp90 was markedly reduced and the inhibitory effects of ATP or 17-AAG were negligible. The dimerization between endogenous Hsp90alpha and exogenous HA-Hsp90beta was confirmed by immunoprecipitation, however the association between eNOS and D88N-Hsp90 was less than WT-Hsp90. Furthermore, adenoviral transduction of bovine aortic ECs with D88N-Hsp90 suppressed VEGF-induced phosphorylation of Akt, eNOS, and NO release and the inhibitory effect was blocked by okadaic acid. Moreover, D88N-Hsp90 abolished VEGF-stimulated Rac activation and suppressed VEGF-induced stress fiber formation. Transduction with D88N-Hsp90 decreased growth medium mediated migration of wild-type ECs, but not Akt1(-/-) ECs suggesting that Akt is key target of Hsp90. CONCLUSIONS: Our data demonstrate that dominant-negative Hsp90 modulates endothelial cell mobility mainly through PP2A-mediated dephosphorylation of Akt and Rac activation.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/enzimologia , Proteínas de Choque Térmico HSP90/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Adenoviridae , Animais , Bovinos , Células Cultivadas , Pulmão/citologia , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais
13.
J Exp Med ; 204(10): 2373-82, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17893196

RESUMO

Caveolin-1 (Cav-1) is the principal structural component of caveolae organelles in smooth muscle cells, adipocytes, fibroblasts, epithelial cells, and endothelial cells (ECs). Cav-1-deficient (Cav-1 knockout [KO]) mice are viable and show increases of nitric oxide (NO) production in vasculature, cardiomyopathy, and pulmonary dysfunction. In this study, we generated EC-specific Cav-1-reconstituted (Cav-1 RC) mice and reexamined vascular, cardiac, and pulmonary phenotypes. Cav-1 KO pulmonary arteries had decreased smooth muscle contractility and increased endothelial NO synthase activation and hypotension; the latter two effects were rescued completely in Cav-1 RC mice. Cav-1 KO mice exhibited myocardial hypertrophy, pulmonary hypertension, and alveolar cell hyperproliferation caused by constitutive activation of p42/44 mitogen-activated protein kinase and Akt. Interestingly, in Cav-1 RC mice, cardiac hypertrophy and pulmonary hypertension were completely rescued, whereas alveolar hyperplasia was partially recovered because of the lack of rescue of Cav-1 in bronchiolar epithelial cells. These results provide clear physiological evidence supporting the important role of cell type-specific Cav-1 expression governing multiple phenotypes in the vasculature, heart, and lung.


Assuntos
Vasos Sanguíneos/metabolismo , Caveolina 1/metabolismo , Regulação da Expressão Gênica , Cardiopatias Congênitas/metabolismo , Pneumopatias/metabolismo , Animais , Vasos Sanguíneos/patologia , Peso Corporal , Caveolina 1/deficiência , Caveolina 1/genética , Colágeno/biossíntese , Endotélio/metabolismo , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Pneumopatias/genética , Pneumopatias/patologia , Camundongos , Camundongos Knockout , Contração Miocárdica , Óxido Nítrico Sintase Tipo III/metabolismo , Fenótipo , Transdução de Sinais
14.
J Biol Chem ; 282(22): 16631-43, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17416589

RESUMO

Various cellular signals initiate calcium entry into cells, and there is evidence that lipid rafts and caveolae may concentrate proteins that regulate transmembrane calcium fluxes. Here, using mice deficient in caveolin-1 (Cav-1) and Cav-1 knock-out reconstituted with endothelium-specific Cav-1, we show that Cav-1 is essential for calcium entry in endothelial cells and governs the localization and protein-protein interactions between transient receptor channels C4 and C1. Thus, Cav-1 is required for calcium entry in vascular endothelial cells and perhaps other specialized cell types containing caveolae.


Assuntos
Cálcio/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Células Endoteliais/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Caveolina 1/genética , Células Endoteliais/ultraestrutura , Transporte de Íons/genética , Camundongos , Camundongos Knockout
15.
Cancer Res ; 67(6): 2849-56, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363608

RESUMO

Caveolin-1 (Cav-1) is a major structural protein that is essential to the formation of the organelle, caveolae. Cav-1 knockout (KO) mice were observed to be completely devoid of caveolae yet they exhibit a hyperpermeable vasculature. Given the nature of the hyperpermeable Cav-1 KO endothelium, we sought to investigate if tumors grown in Cav-1 KO mice would be leaky and grow faster. Indeed, Lewis lung carcinoma cells implanted into Cav-1 KO mice had increased tumor vascular permeability, measured by Evans blue extravasation and fibrinogen deposition compared with tumors implanted into wild-type (WT) mice. Cav-1 KO mice also had significantly higher tumor growth rates, attributable to increased tumor angiogenesis and decreased tumor cell death. Furthermore, administration of an antipermeability peptide, cavtratin, was able to correct the tumor hyperpermeability as well as attenuate the increased tumor growth. Mechanistically, endothelial cells isolated from Cav-1 KO mice exhibited increased tyrosine phosphorylation on vascular endothelial growth factor (VEGF) receptor-2 (VEGFR-2) and decreased association with the adherens junction protein, VE-cadherin. Thus, the loss of Cav-1 increases tumor permeability and growth and that may relate to enhanced VEGF signaling due to lack of Cav-1 inhibition of VEGFR-2 or decreased VE-cadherin mediated VEGFR-2 phosphorylation.


Assuntos
Carcinoma Pulmonar de Lewis/irrigação sanguínea , Caveolina 1/deficiência , Junções Aderentes/metabolismo , Sequência de Aminoácidos , Animais , Apoptose/fisiologia , Permeabilidade Capilar , Carcinoma Pulmonar de Lewis/patologia , Caveolina 1/genética , Processos de Crescimento Celular/fisiologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fosforilação , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Cancer Cell ; 10(2): 159-70, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16904613

RESUMO

Endothelial cells in growing tumors express activated Akt, which when modeled by transgenic endothelial expression of myrAkt1 was sufficient to recapitulate the abnormal structural and functional features of tumor blood vessels in nontumor tissues. Sustained endothelial Akt activation caused increased blood vessel size and generalized edema from chronic vascular permeability, while acute permeability in response to VEGF-A was unaffected. These changes were reversible, demonstrating an ongoing requirement for Akt signaling for the maintenance of these phenotypes. Furthermore, rapamycin inhibited endothelial Akt signaling, vascular changes from myrAkt1, tumor growth, and tumor vascular permeability. Akt signaling in the tumor vascular stroma was sensitive to rapamycin, suggesting that rapamycin may affect tumor growth in part by acting as a vascular Akt inhibitor.


Assuntos
Células Endoteliais/patologia , Endotélio Vascular/patologia , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirolimo/farmacologia , Animais , Permeabilidade Capilar , Células Cultivadas , Edema/metabolismo , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/fisiologia
18.
J Cell Biol ; 174(3): 369-77, 2006 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-16864653

RESUMO

Lipid modifications mediate the subcellular localization and biological activity of many proteins, including endothelial nitric oxide synthase (eNOS). This enzyme resides on the cytoplasmic aspect of the Golgi apparatus and in caveolae and is dually acylated by both N-myristoylation and S-palmitoylation. Palmitoylation-deficient mutants of eNOS release less nitric oxide (NO). We identify enzymes that palmitoylate eNOS in vivo. Transfection of human embryonic kidney 293 cells with the complementary DNA (cDNA) for eNOS and 23 cDNA clones encoding the Asp-His-His-Cys motif (DHHC) palmitoyl transferase family members showed that five clones (2, 3, 7, 8, and 21) enhanced incorporation of [3H]-palmitate into eNOS. Human endothelial cells express all five of these enzymes, which colocalize with eNOS in the Golgi and plasma membrane and interact with eNOS. Importantly, inhibition of DHHC-21 palmitoyl transferase, but not DHHC-3, in human endothelial cells reduces eNOS palmitoylation, eNOS targeting, and stimulated NO production. Collectively, our data describe five new Golgi-targeted DHHC enzymes in human endothelial cells and suggest a regulatory role of DHHC-21 in governing eNOS localization and function.


Assuntos
Aciltransferases/metabolismo , Complexo de Golgi/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Ácido Palmítico/metabolismo , Acilação , Motivos de Aminoácidos , Animais , Células COS , Membrana Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Humanos , Imunoprecipitação , Óxido Nítrico/metabolismo , Ligação Proteica , Transporte Proteico
19.
Proc Natl Acad Sci U S A ; 103(29): 11021-6, 2006 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16832062

RESUMO

VEGF, nitric oxide (NO), inflammation, and vascular- and extravascular remodeling coexist in asthma and other disorders. In these responses, VEGF regulates angiogenesis. VEGF also induces inflammation and remodeling. The mechanisms of the latter responses have not been defined, however. We hypothesized that VEGF-induces extravascular tissue responses via NO-dependent mechanisms. To evaluate this hypothesis, we compared the effects of transgenic VEGF165 in lungs from normal mice, mice treated with pan-NO synthase (NOS) or endothelial NOS (eNOS) inhibitors, and mice with null mutations of inducible NOS (iNOS) or eNOS. These studies demonstrate that VEGF selectively stimulates eNOS and iNOS. They also demonstrate that VEGF induces pulmonary alterations via NO-dependent and -independent mechanisms with angiogenesis, edema, mucus metaplasia, airway hyperresponsiveness, lymphocyte accumulation, dendritic cell hyperplasia and S-nitrosoglutathione reductase stimulation being NO-dependent and dendritic cell activation being NO-independent. Furthermore, they demonstrate that eNOS and iNOS both contribute to these responses. NO/NOS-based interventions may be therapeutic in VEGF-driven inflammation and remodeling.


Assuntos
Asma/metabolismo , Asma/patologia , Glândulas Exócrinas/metabolismo , Pulmão/irrigação sanguínea , Muco/metabolismo , Óxido Nítrico/metabolismo , Fatores de Crescimento do Endotélio Vascular/farmacologia , Álcool Desidrogenase , Animais , Asma/induzido quimicamente , Asma/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Glutationa Redutase/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Metaplasia/induzido quimicamente , Metaplasia/complicações , Metaplasia/metabolismo , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Óxido Nítrico Sintase/metabolismo , Fatores de Crescimento do Endotélio Vascular/biossíntese , Fatores de Crescimento do Endotélio Vascular/genética
20.
J Clin Invest ; 116(5): 1284-91, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16670769

RESUMO

Caveolae in endothelial cells have been implicated as plasma membrane microdomains that sense or transduce hemodynamic changes into biochemical signals that regulate vascular function. Therefore we compared long- and short-term flow-mediated mechanotransduction in vessels from WT mice, caveolin-1 knockout (Cav-1 KO) mice, and Cav-1 KO mice reconstituted with a transgene expressing Cav-1 specifically in endothelial cells (Cav-1 RC mice). Arterial remodeling during chronic changes in flow and shear stress were initially examined in these mice. Ligation of the left external carotid for 14 days to lower blood flow in the common carotid artery reduced the lumen diameter of carotid arteries from WT and Cav-1 RC mice. In Cav-1 KO mice, the decrease in blood flow did not reduce the lumen diameter but paradoxically increased wall thickness and cellular proliferation. In addition, in isolated pressurized carotid arteries, flow-mediated dilation was markedly reduced in Cav-1 KO arteries compared with those of WT mice. This impairment in response to flow was rescued by reconstituting Cav-1 into the endothelium. In conclusion, these results showed that endothelial Cav-1 and caveolae are necessary for both rapid and long-term mechanotransduction in intact blood vessels.


Assuntos
Cavéolas/metabolismo , Caveolina 1/fisiologia , Endotélio Vascular/patologia , Animais , Artérias Carótidas/patologia , Caveolina 1/genética , Células Endoteliais , Endotélio Vascular/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Transdução de Sinais , Estresse Mecânico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA