Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 18(3): 250-251, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28198829
2.
PLoS Biol ; 17(4): e3000209, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31017904

RESUMO

Gram+ infections are worldwide life-threatening diseases in which the pathological role of type I interferon (IFN) has been highlighted. Plasmacytoid predendritic cells (pDCs) produce high amounts of type I IFN following viral sensing. Despite studies suggesting that pDCs respond to bacteria, the mechanisms underlying bacterial sensing in pDCs are unknown. We show here that human primary pDCs express toll-like receptor 1 (TLR1) and 2 (TLR2) and respond to bacterial lipoproteins. We demonstrated that pDCs differentially respond to gram+ bacteria through the TLR1/2 pathway. Notably, up-regulation of costimulatory molecules and pro-inflammatory cytokines was TLR1 dependent, whereas type I IFN secretion was TLR2 dependent. Mechanistically, we demonstrated that these differences relied on diverse signaling pathways activated by TLR1/2. MAPK and NF-κB pathways were engaged by TLR1, whereas the Phosphoinositide 3-kinase (PI3K) pathway was activated by TLR2. This dichotomy was reflected in a different role of TLR2 and TLR1 in pDC priming of naïve cluster of differentiation 4+ (CD4+) T cells, and T helper (Th) cell differentiation. This work provides the rationale to explore and target pDCs in bacterial infection.


Assuntos
Células Dendríticas/metabolismo , Infecções por Bactérias Gram-Positivas/metabolismo , Receptor 1 Toll-Like/metabolismo , Receptor 2 Toll-Like/metabolismo , Diferenciação Celular/fisiologia , Citocinas/metabolismo , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Infecções por Bactérias Gram-Positivas/patologia , Voluntários Saudáveis , Humanos , Interferon-alfa/metabolismo , Ativação Linfocitária , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Linfócitos T/imunologia
4.
J Allergy Clin Immunol ; 134(2): 373-81, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24910175

RESUMO

BACKGROUND: Thymic stromal lymphopoietin (TSLP) is a major proallergic cytokine that promotes TH2 responses through dendritic cell (DC) activation. Whether it also plays a role in human autoimmune inflammation and associated pathways is not known. OBJECTIVE: In this study we investigated the potential role of several epithelium-derived factors, including TSLP, in inducing IL-23 production by human DCs. We further dissected the role of TSLP in patients with psoriasis, an IL-23-associated skin autoimmune disease. METHODS: The study was performed in human subjects using primary cells and tissue samples from patients with psoriasis and healthy donors. We analyzed the production of IL-23 in vitro by blood and skin DCs. We studied the function for TSLP and its interaction with other components of the inflammatory microenvironment in situ and ex vivo. RESULTS: We found that TSLP synergized with CD40 ligand to promote DC activation and pathogenic IL-23 production by primary blood and skin DCs. In situ TSLP was strongly expressed by keratinocytes of untreated psoriatic lesions but not in normal skin. Moreover, we could demonstrate that IL-4, an important component of the TH2 inflammation seen in patients with atopic dermatitis, inhibited IL-23 production induced by TSLP and CD40 ligand in a signal transducer and activator of transcription 6-independent manner. CONCLUSION: Our results identify TSLP as a novel player within the complex psoriasis cytokine network. Blocking TSLP in patients with psoriasis might contribute to decreasing DC activation and shutting down the production of pathogenic IL-23.


Assuntos
Citocinas/imunologia , Células Dendríticas/imunologia , Interleucina-23/imunologia , Queratinócitos/imunologia , Psoríase/imunologia , Pele/imunologia , Adulto , Ligante de CD40/genética , Ligante de CD40/imunologia , Citocinas/genética , Células Dendríticas/patologia , Dermatite Atópica/genética , Dermatite Atópica/imunologia , Dermatite Atópica/patologia , Regulação da Expressão Gênica , Humanos , Interleucina-23/genética , Interleucina-4/genética , Interleucina-4/imunologia , Queratinócitos/patologia , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , Psoríase/genética , Psoríase/patologia , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/imunologia , Transdução de Sinais , Pele/patologia , Células Th2/imunologia , Células Th2/patologia , Linfopoietina do Estroma do Timo
5.
Clin Cancer Res ; 28(15): 3387-3399, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35121624

RESUMO

PURPOSE: While patients responding to checkpoint blockade often achieve remarkable clinical responses, there is still significant unmet need due to resistant or refractory tumors. A combination of checkpoint blockade with further T-cell stimulation mediated by 4-1BB agonism may increase response rates and durability of response. A bispecific molecule that blocks the programmed cell death 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) axis and localizes 4-1BB costimulation to a PD-L1-positive (PD-L1+) tumor microenvironment (TME) or tumor draining lymph nodes could maximize antitumor immunity and increase the therapeutic window beyond what has been reported for anti-4-1BB mAbs. EXPERIMENTAL DESIGN: We generated and characterized the PD-L1/4-1BB bispecific molecule PRS-344/S095012 for target binding and functional activity in multiple relevant in vitro assays. Transgenic mice expressing human 4-1BB were transplanted with human PD-L1-expressing murine MC38 cells to assess in vivo antitumoral activity. RESULTS: PRS-344/S095012 bound to its targets with high affinity and efficiently blocked the PD-1/PD-L1 pathway, and PRS-344/S095012-mediated 4-1BB costimulation was strictly PD-L1 dependent. We demonstrated a synergistic effect of both pathways on T-cell stimulation with the bispecific PRS-344/S095012 being more potent than the combination of mAbs. PRS-344/S095012 augmented CD4-positive (CD4+) and CD8-positive (CD8+) T-cell effector functions and enhanced antigen-specific T-cell stimulation. Finally, PRS-344/S095012 demonstrated strong antitumoral efficacy in an anti-PD-L1-resistant mouse model in which soluble 4-1BB was detected as an early marker for 4-1BB agonist activity. CONCLUSIONS: The PD-L1/4-1BB bispecific PRS-344/S095012 efficiently combines checkpoint blockade with a tumor-localized 4-1BB-mediated stimulation burst to antigen-specific T cells, more potent than the combination of mAbs, supporting the advancement of PRS-344/S095012 toward clinical development. See related commentary by Shu et al., p. 3182.


Assuntos
Antígeno B7-H1 , Neoplasias , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/imunologia , Humanos , Fatores Imunológicos/uso terapêutico , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/imunologia , Microambiente Tumoral
6.
J Clin Invest ; 118(6): 2062-75, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18483621

RESUMO

Experimental and clinical evidence indicate that bone marrow cells participate in the process of new blood vessel formation. However, the molecular mechanisms underlying their recruitment and their exact role are still elusive. Here, we show that bone marrow cells are recruited to the sites of neoangiogenesis through the neuropilin-1 (NP-1) receptor and that they are essential for the maturation of the activated endothelium and the formation of arteries in mice. By exploiting adeno-associated virus vector-mediated, long-term in vivo gene expression, we show that the 165-aa isoform of VEGF, which both activates the endothelium and recruits NP-1+ myeloid cells, is a powerful arteriogenic agent. In contrast, neither the shortest VEGF121 isoform, which does not bind NP-1 and thus does not recruit bone marrow cells, nor semaphorin 3A, which attracts cells but inhibits endothelial activation, are capable of sustaining arterial formation. Bone marrow myeloid cells are not arteriogenic per se nor are they directly incorporated in the newly formed vasculature, but they contribute to arterial formation through a paracrine effect ensuing in the activation and proliferation of tissue-resident smooth muscle cells.


Assuntos
Artérias/patologia , Células da Medula Óssea/citologia , Neuropilina-1/fisiologia , Animais , Células da Medula Óssea/metabolismo , Antígeno CD11b/biossíntese , Proliferação de Células , Dependovirus/metabolismo , Regulação da Expressão Gênica , Terapia Genética/métodos , Camundongos , Neovascularização Patológica , Neuropilina-1/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Semaforina-3A/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
FASEB J ; 24(5): 1467-78, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20019242

RESUMO

Mounting evidence indicates that the function of members of the vascular endothelial growth factor (VEGF) family extends beyond blood vessel formation. Here, we show that the prolonged intramyocardial expression of VEGF-A(165) and VEGF-B(167) on adeno-associated virus-mediated gene delivery determined a marked improvement in cardiac function after myocardial infarction in rats, by promoting cardiac contractility, preserving viable cardiac tissue, and preventing remodeling of the left ventricle (LV) over time. Consistent with this functional outcome, animals treated with both factors showed diminished fibrosis and increased contractile myocardium, which were more pronounced after expression of the selective VEGF receptor-1 (VEGFR-1) ligand VEGF-B, in the absence of significant induction of angiogenesis. We found that cardiomyocytes expressed VEGFR-1, VEGFR-2, and neuropilin-1 and that, in particular, VEGFR-1 was specifically up-regulated in hypoxia and on exposure to oxidative stress. VEGF-B exerted powerful antiapoptotic effect in both cultured cardiomyocytes and after myocardial infarction in vivo. Finally, VEGFR-1 activation by VEGF-B was found to elicit a peculiar gene expression profile proper of the compensatory, hypertrophic response, consisting in activation of alphaMHC and repression of betaMHC and skeletal alpha-actin, and an increase in SERCA2a, RYR, PGC1alpha, and cardiac natriuretic peptide transcripts, both in cultured cardiomyocytes and in infarcted hearts. The finding that VEGFR-1 activation by VEGF-B prevents loss of cardiac mass and promotes maintenance of cardiac contractility over time has obvious therapeutic implications.


Assuntos
Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/agonistas , Animais , Apoptose/genética , Células Cultivadas , Humanos , Masculino , Contração Miocárdica/genética , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Neovascularização Fisiológica/genética , Ratos , Ratos Wistar , Ativação Transcricional , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/genética , Remodelação Ventricular/genética
8.
Hum Gene Ther ; 18(6): 515-24, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17559317

RESUMO

Although the angiogenic effect of vascular endothelial growth factor (VEGF) is widely recognized, a central question concerns whether the vessels formed on its overexpression effectively increase tissue perfusion in vivo. To explore this issue, here we exploit AAV vectors to obtain the prolonged expression of VEGF and angiopoietin-1 (Ang1) in rat skeletal muscle. Over a period of 6 months, muscle blood flow (MBF) and vascular permeability were measured by positron emission tomography and single-photon emission computed tomography, respectively. All measurements were performed under resting conditions and after electrically induced muscle exercise. Despite the potent angiogenic effect of VEGF, documented by vessel counting and intravascular volume assessment, the expression of this factor did not improve resting MBF, and it even decreased perfusion after exercise. This deleterious effect was related to the formation of leaky vascular lacunae, which accounted for the occurrence of arteriovenous shunts that excluded the downstream microcirculation. These effects were significantly counteracted by the coinjection of VEGF and Ang1, which determined a marked increase in resting MBF and, most notably, a significant improvement after exercise that persisted over time. Taken together, these results challenge the effectiveness of VEGF as a sole factor to induce angiogenesis and suggest the use of factor combinations to achieve competent vessel formation.


Assuntos
Angiopoietina-1/análogos & derivados , Endotélio Vascular/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Patológica/diagnóstico por imagem , Fator A de Crescimento do Endotélio Vascular/genética , Angiopoietina-1/genética , Animais , Permeabilidade Capilar , Dependovirus/genética , Endotélio Vascular/patologia , Expressão Gênica/fisiologia , Vetores Genéticos , Masculino , Modelos Animais , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Ratos , Ratos Wistar , Pentetato de Tecnécio Tc 99m , Tomografia Computadorizada de Emissão de Fóton Único , Transfecção
9.
J Exp Med ; 214(5): 1529-1546, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28428203

RESUMO

T follicular helper cells (Tfh) are important regulators of humoral responses. Human Tfh polarization pathways have been thus far associated with Th1 and Th17 polarization pathways. How human Tfh cells differentiate in Th2-skewed environments is unknown. We show that thymic stromal lymphopoietin (TSLP)-activated dendritic cells (DCs) promote human Tfh differentiation from naive CD4 T cells. We identified a novel population, distinct from Th2 cells, expressing IL-21 and TNF, suggestive of inflammatory cells. TSLP-induced T cells expressed CXCR5, CXCL13, ICOS, PD1, BCL6, BTLA, and SAP, among other Tfh markers. Functionally, TSLP-DC-polarized T cells induced IgE secretion by memory B cells, and this depended on IL-4Rα. TSLP-activated DCs stimulated circulating memory Tfh cells to produce IL-21 and CXCL13. Mechanistically, TSLP-induced Tfh differentiation depended on OX40-ligand, but not on ICOS-ligand. Our results delineate a pathway of human Tfh differentiation in Th2 environments.


Assuntos
Citocinas/fisiologia , Células Dendríticas/fisiologia , Ligante OX40/fisiologia , Células Th2/fisiologia , Diferenciação Celular/fisiologia , Quimiocina CXCL13/metabolismo , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interleucinas/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Receptores CXCR5/metabolismo , Receptores Imunológicos/metabolismo , Linfopoietina do Estroma do Timo
10.
Methods Mol Biol ; 1423: 153-67, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27142015

RESUMO

Blood represents the most accessible source of human dendritic cells (DCs). We present here a method to isolate three DC subtypes, as identified until now, from peripheral blood: plasmacytoid dendritic cells (pDCs), CD141(+) myeloid DCs, and CD1c(+) myeloid DCs. The method is based on the sequential depletion of non-DCs. First, depletion of granulocytes, erythrocytes, and platelets is obtained by blood centrifugation over a Ficoll gradient. Then, antibodies recognizing non-DCs, combined with magnetic beads, allow enrichment of DCs from peripheral blood mononuclear cells (PBMCs). Finally, enriched DCs are purified and separated into the different subtypes by immunolabeling and fluorescence-activated cell sorting (FACS) using DC-specific surface markers.DC studies might contribute to the comprehension of human immune processes in physiological and pathological conditions. Human blood DCs targeting might be a useful tool to ameliorate inflammatory diseases and improve vaccination strategies.


Assuntos
Separação Celular/métodos , Células Dendríticas/citologia , Leucócitos Mononucleares/citologia , Antígenos CD1/metabolismo , Antígenos de Superfície/metabolismo , Células Dendríticas/imunologia , Citometria de Fluxo , Glicoproteínas/metabolismo , Humanos , Leucócitos Mononucleares/imunologia , Trombomodulina
11.
Oncoimmunology ; 5(8): e1178438, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27622057

RESUMO

Thymic stromal lymphopoietin (TSLP) is an epithelial cell-derived cytokine that primes dendritic cells for Th2 induction. It has been implicated in different types of allergic diseases. Recent work suggested that TSLP could play an important role in the tumor microenvironment and influence tumor progression, in particular in breast cancer. In this study we systematically assessed the production of TSLP at the mRNA and protein levels in several human breast cancer cell lines, large-scale public transcriptomics data sets, and primary human breast tumors. We found that TSLP production was marginal, and concerned less than 10% of the tumors, with very low mRNA and protein levels. In most cases TSLP was undetectable and found to be expressed at lower levels in breast cancer as compared to normal breast tissue. Last, we could not detect any functional TSLP receptor (TSLPR) expression neither on hematopoietic cells nor on stromal cells within the primary tumor microenvironment. We conclude that TSLP-TSLPR pathway activity is not significantly detected within human breast cancer. Taken together, these observations do not support TSLP targeting in breast cancer.

12.
Oncoimmunology ; 5(7): e1179414, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27622034

RESUMO

Thymic stromal lymphopoietin (TSLP) is an interleukin (IL)-7-like cytokine expressed by epithelial cells during allergic inflammation, and activating dendritic cells (DC). Its expression and functional role in cancer remain controversial. We conducted retrospective (n = 89), and prospective studies including patients with untreated primary head and neck squamous cell carcinoma (HNSCC). We found that TSLP was overexpressed by HNSCC tumor cells, and associated with a highly differentiated status. However, no significant difference in overall and recurrence-free survival was found between patients bearing a tumor with high and low TSLP levels, respectively. Surprisingly, there was no significant association between the levels of TSLP expression, and the number of tumor-infiltrating mature DCLAMP(+) DC. In order to explain the apparent lack of TSLP-induced DC activation, we performed phenotypic and functional experiments on freshly resected tumors. Tumor-infiltrating immune cells, including DC, did not express the TSLP receptor heterodimer (TSLPR chain, IL-7Ralpha chain). Furthermore, freshly sorted blood CD11c(+) DC from healthy donors cultured with tumor-conditioned supernatant exhibited an activated profile, but this was not affected by an anti-TSLP blocking antibody, suggesting a DC activation pathway independent of tumor-derived TSLP. Overall, our results demonstrate that TSLP is overexpressed in HNSCC but its function is hampered by the lack of TSLPR-expressing cells in the tumor microenvironment. Such a dissociated ligand-receptor expression may impact intercellular communication in other immune activation pathways, and tumor types.

13.
Curr Opin Immunol ; 32: 42-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25588554

RESUMO

Innate immune cells are generated through central and peripheral differentiation pathways, and receive multiple signals from tissue microenvironment. The complex interplay between immune cell state and environmental signals is crucial for the adaptation and efficient response to pathogenic threats. Here, we discuss how systems biology approaches have brought global view and high resolution to the characterization of (1) immune cell diversity, (2) phenotypic, transcriptional and functional changes in response to environmental signals, (3) integration of multiple stimuli. We will mostly focus on systems level studies in dendritic cells and macrophages. Generalization of these approaches should elucidate innate immune cell diversity and plasticity, and may be used in the human to generate hypothesis on cell filiation and novel strategies for immunotherapy.


Assuntos
Sistema Imunitário/citologia , Sistema Imunitário/fisiologia , Imunidade Inata , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo
14.
J Tissue Eng ; 6: 2041731415611717, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26848383

RESUMO

In regenerative medicine, new approaches are required for the creation of tissue substitutes, and the interplay between different research areas, such as tissue engineering, microsurgery and gene therapy, is mandatory. In this article, we report a modification of a published model of tissue engineering, based on an arterio-venous loop enveloped in a cross-linked collagen-glycosaminoglycan template, which acts as an isolated chamber for angiogenesis and new tissue formation. In order to foster tissue formation within the chamber, which entails on the development of new vessels, we wondered whether we might combine tissue engineering with a gene therapy approach. Based on the well-described tropism of adeno-associated viral vectors for post-mitotic tissues, a muscular flap was harvested from the pectineus muscle, inserted into the chamber and transduced by either AAV vector encoding human VEGF165 or AAV vector expressing the reporter gene ß-galactosidase, as a control. Histological analysis of the specimens showed that muscle transduction by AAV vector encoding human VEGF165 resulted in enhanced tissue formation, with a significant increase in the number of arterioles within the chamber in comparison with the previously published model. Pectineus muscular flap, transduced by adeno-associated viral vectors, acted as a source of the proangiogenic factor vascular endothelial growth factor, thus inducing a consistent enhancement of vessel growth into the newly formed tissue within the chamber. In conclusion, our present findings combine three different research fields such as microsurgery, tissue engineering and gene therapy, suggesting and showing the feasibility of a mixed approach for regenerative medicine.

15.
J Mol Cell Biol ; 6(2): 116-27, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24620033

RESUMO

The tyrosine kinase receptor vascular endothelial growth factor receptor 2 (VEGFR2) is a key regulator of angiogenesis. Here we show that VEGFR2 is acetylated in endothelial cells both at four lysine residues forming a dense cluster in the kinase insert domain and at a single lysine located in the receptor activation loop. These modifications are under dynamic control of the acetyltransferase p300 and two deacetylases HDAC5 and HDAC6. We demonstrate that VEGFR2 acetylation essentially regulates receptor phosphorylation. In particular, VEGFR2 acetylation significantly alters the kinetics of receptor phosphorylation after ligand binding, allowing receptor phosphorylation and intracellular signaling upon prolonged stimulation with VEGF. Molecular dynamics simulations indicate that acetylation of the lysine in the activation loop contributes to the transition to an open active state, in which tyrosine phosphorylation is favored by better exposure of the kinase target residues. These findings indicate that post-translational modification by acetylation is a critical mechanism that directly affects VEGFR2 function.


Assuntos
Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Proteína p300 Associada a E1A/metabolismo , Células HEK293 , Histona Desacetilases/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligantes , Lisina/metabolismo , Espectrometria de Massas , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Estabilidade Proteica , Estrutura Secundária de Proteína , Sus scrofa , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química
16.
Cancer Res ; 72(24): 6371-81, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23222303

RESUMO

Improving tumor perfusion, thus tempering tumor-associated hypoxia, is known to impair cancer progression. Previous work from our laboratory has shown that VEGF-A165 and semaphorin 3A (Sema3A) promote vessel maturation through the recruitment of a population of circulating monocytes expressing the neuropilin-1 (Nrp1) receptor (Nrp1-expressing monocytes; NEM). Here, we define the characteristics of bone marrow NEMs and assess whether these cells might represent an exploitable tool to induce tumor vessel maturation. Gene expression signature and surface marker analysis have indicated that NEMs represent a specific subset of CD11b+ Nrp1+ Gr1- resident monocytes, distinctively recruited by Sema3A. NEMs were found to produce several factors involved in vessel maturation, including PDGFb, TGF-ß, thrombospondin-1, and CXCL10; consistently, they were chemoattractive for vascular smooth muscle cells in vitro. When directly injected into growing tumors, NEMs, isolated either from the bone marrow or from Sema3A-expressing muscles, exerted antitumor activity despite having no direct effects on the proliferation of tumor cells. NEM inoculation specifically promoted mural cell coverage of tumor vessels and decreased vascular leakiness. Tumors treated with NEMs were smaller, better perfused and less hypoxic, and had a reduced level of activation of HIF-1α. We conclude that NEMs represent a novel, unique population of myeloid cells that, once inoculated into a tumor, induce tumor vessel normalization and inhibit tumor growth.


Assuntos
Proliferação de Células , Monócitos/fisiologia , Neoplasias/terapia , Neovascularização Patológica/prevenção & controle , Neuropilina-1/fisiologia , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/fisiologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/fisiologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/terapia , Neuropilina-1/genética , Neuropilina-1/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/fisiologia
17.
J Mol Med (Berl) ; 86(1): 105-15, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17957349

RESUMO

Skeletal myogenesis is a multistep process starting with progenitor cell proliferation, followed by their exit from the cell cycle, differentiation, alignment, and fusion to form multinucleated myotubes, typical of the differentiated muscle tissue. While the molecular players involved in early myogenesis have been extensively characterized, information about the later steps of the process is scanty. Here, we describe a novel myogenic cell line (MYOP7), composed of highly proliferating Sca-1+ muscle precursor cells, which can be induced to terminally differentiate into spontaneously contracting multinucleated myotubes. By performing high-density microarray analysis on these cells, we identified a series of genes, differentially expressed in proliferating vs differentiating conditions, which are candidates to play a major role in the later phase of myogenesis. In addition, we confirmed that the late stages of muscle differentiation are characterized by a marked upregulation of the cellular receptors for the vascular endothelial growth factor.


Assuntos
Linhagem Celular , Desenvolvimento Muscular , Músculo Esquelético/citologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Diferenciação Celular , Camundongos , Músculo Esquelético/fisiologia , Receptores de Superfície Celular/genética , Células-Tronco/citologia , Regulação para Cima
18.
Microsurgery ; 27(7): 623-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17868145

RESUMO

BACKGROUND: Microsurgical tissue engineering is an emerging topic in regenerative medicine. Here we describe a new microsurgical model of bioengineering in rats based on the use of an arterovenous loop (AV) implanted into a commercially available crosslinked collagen/glycosaminoglycan template. METHODS: The microvascular loop was created between the femoral artery and vein and covered by the template folded onto itself. The chamber was isolated from the outside tissue by an outer silicon layer to impede tissue ingrowth. RESULTS: At 1-month postimplantation, the tissue chamber was found heavily vascularized, as assessed by laser Doppler perfusion analysis. Histological examination showed that the AV loop was integrated into the collagen matrix of the template and that the whole template was filled with a newly formed soft connective tissue. Most interestingly, the whole scaffold was found heavily vascularized, including the formation of a large number of alpha-SMA-positive arterioles. CONCLUSIONS: The developed microsurgical chamber provides a highly vascular, isolated tool for in vivo tissue engineering.


Assuntos
Artéria Femoral/cirurgia , Veia Femoral/cirurgia , Microcirurgia , Engenharia Tecidual/métodos , Anastomose Cirúrgica , Animais , Colágeno , Reagentes de Ligações Cruzadas , Cultura em Câmaras de Difusão , Glicosaminoglicanos , Masculino , Ratos , Ratos Wistar , Engenharia Tecidual/instrumentação
19.
Cell ; 131(3): 463-75, 2007 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17981115

RESUMO

Novel antiangiogenic strategies with complementary mechanisms are needed to maximize efficacy and minimize resistance to current angiogenesis inhibitors. We explored the therapeutic potential and mechanisms of alphaPlGF, an antibody against placental growth factor (PlGF), a VEGF homolog, which regulates the angiogenic switch in disease, but not in health. alphaPlGF inhibited growth and metastasis of various tumors, including those resistant to VEGF(R) inhibitors (VEGF(R)Is), and enhanced the efficacy of chemotherapy and VEGF(R)Is. alphaPlGF inhibited angiogenesis, lymphangiogenesis, and tumor cell motility. Distinct from VEGF(R)Is, alphaPlGF prevented infiltration of angiogenic macrophages and severe tumor hypoxia, and thus, did not switch on the angiogenic rescue program responsible for resistance to VEGF(R)Is. Moreover, it did not cause or enhance VEGF(R)I-related side effects. The efficacy and safety of alphaPlGF, its pleiotropic and complementary mechanism to VEGF(R)Is, and the negligible induction of an angiogenic rescue program suggest that alphaPlGF may constitute a novel approach for cancer treatment.


Assuntos
Anticorpos Monoclonais/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteínas da Gravidez/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais/efeitos adversos , Antineoplásicos/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Saúde , Humanos , Linfangiogênese/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Fator de Crescimento Placentário , Resultado do Tratamento
20.
Blood ; 107(9): 3546-54, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16391016

RESUMO

Vascular endothelial growth factor (VEGF) is a key regulator of blood vessel formation during both vasculogenesis and angiogenesis. The prolonged expression of VEGF in the normoperfused skeletal muscles of adult rodents after gene transfer using AAV vectors induces the formation of a large set of new capillaries and small arteries. Notably, this process is accompanied by the massive infiltration by mononuclear cells. This observation raises the possibility that these cells might represent circulating progenitors that are eventually incorporated in the newly formed vessels. Here we explore this possibility by exploiting 4 different experimental models based on (a) the transplantation of male bone marrow into female recipients; (b) the transplantation of Tie2-GFP transgenic bone marrow; (c) the transplantation of bone marrow in the presence of erythropoietin (EPO), a mobilizer of endothelial progenitor cells (EPCs); and (d) the reimplantation of ex vivo-expanded EPCs. In all 4 models, VEGF acted as a powerful attractor of bone marrow-derived mononuclear cells, bearing different myeloid and endothelial markers proper of the EPCs to the sites of neovascularization. In no case, however, were the attracted cells incorporated in the newly formed vasculature. These observations indicate that new blood vessel formation induced by VEGF occurs through bona fide sprouting angiogenesis; the contribution of the infiltrating bone marrow-derived cells to this process still remains enigmatic.


Assuntos
Células da Medula Óssea/fisiologia , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Sequência de Bases , Vasos Sanguíneos/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Transplante de Medula Óssea , Diferenciação Celular , Movimento Celular , DNA/genética , Dependovirus/genética , Eritropoetina/farmacologia , Feminino , Expressão Gênica , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Músculo Esquelético/irrigação sanguínea , Proteínas Recombinantes , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA