Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 14: 1109452, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37064885

RESUMO

Introduction: Ventilator-induced lung injury (VILI) may aggravate critical illness. Although angiotensin-converting enzyme (ACE) inhibition has beneficial effects in ventilator-induced lung injury, its clinical application is impeded by concomitant hypotension. We hypothesized that the aminopeptidase inhibitor ALT-00 may oppose the hypotension induced by an angiotensin-converting enzyme inhibitor, and that this combination would activate the alternative renin-angiotensin system (RAS) axis to counteract ventilator-induced lung injury. Methods: In separate experiments, C57BL/6 mice were mechanically ventilated with low (LVT, 6 mL/kg) and high tidal volumes (HVT, 30 mL/kg) for 4 h or remained unventilated (sham). High tidal volume-ventilated mice were treated with lisinopril (0.15 µg/kg/min) ± ALT-00 at 2.7, 10 or 100 µg/kg/min. Blood pressure was recorded at baseline and after 4 h. Lung histology was evaluated for ventilator-induced lung injury and the angiotensin (Ang) metabolite profile in plasma (equilibrium levels of Ang I, Ang II, Ang III, Ang IV, Ang 1-7, and Ang 1-5) was measured with liquid chromatography tandem mass spectrometry at the end of the experiment. Angiotensin concentration-based markers for renin, angiotensin-converting enzyme and alternative renin-angiotensin system activities were calculated. Results: High tidal volume-ventilated mice treated with lisinopril showed a significant drop in the mean arterial pressure at 4 h compared to baseline, which was prevented by adding ALT-00 at 10 and 100 µg/kg/min. Ang I, Ang II and Ang 1-7 plasma equilibrium levels were elevated in the high tidal volumes group versus the sham group. Lisinopril reduced Ang II and slightly increased Ang I and Ang 1-7 levels versus the untreated high tidal volumes group. Adding ALT-00 at 10 and 100 µg/kg/min increased Ang I and Ang 1-7 levels versus the high tidal volume group, and partly prevented the downregulation of Ang II levels caused by lisinopril. The histological lung injury score was higher in the high tidal volume group versus the sham and low tidal volume groups, and was attenuated by lisinopril ± ALT-00 at all dose levels. Conclusion: Combined angiotensin-converting enzyme plus aminopeptidase inhibition prevented systemic hypotension and maintained the protective effect of lisinopril. In this study, a combination of lisinopril and ALT-00 at 10 µg/kg/min appeared to be the optimal approach, which may represent a promising strategy to counteract ventilator-induced lung injury that merits further exploration.

2.
J Immunol Methods ; 449: 28-36, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28647457

RESUMO

Targeting plasma IgE by therapeutic mABs like Omalizumab (Xolair®) is current clinical practice for severe allergic conditions or other IgE related diseases like chronic urticaria. As an alternative to soluble IgE targeting, IgE supply can be lowered by targeting the Extracellular Membrane Proximal Domain (EMPD) of the IgE B cell receptor (BCR) present on IgE switched B cells. This ultimately leads to apoptosis of these cells upon IgE BCR crosslinking. Since tools to selectively assess the efficacy of IgE BCR crosslinking by IgE targeting antibodies are limited, a readily quantifiable cell model was developed that allows to specifically address IgE BCR crosslinking activity in vitro. The new cell model allowed for a direct quantitative comparison of anti-EMPD IgE therapeutic prototype antibody 47H4 with anti-IgE(Ce3) directed therapeutic antibody Omalizumab and with a newly selected anti-human EMPD IgE monoclonal antibody, designated mAB 15cl12. Furthermore, a complementing mouse model was developed that allows for in vivo validation of antibodies addressing human EMPD IgE. It carries a targetable humanized EMPD IgE sequence that has been introduced by seamless genomic replacement of the endogenous EMPD encoding sequence. The model allowed to directly compare IgE lowering activity of two anti-human EMPD IgE therapeutic antibodies in vivo. Our tools provide the means for quantitative assessment of IgE BCR crosslinking activity which is increasingly gaining attention with respect to forthcoming second generation anti-IgE clinical candidates such as Ligelizumab or other clinical candidates featuring additional effector functions such as IgE BCR crosslinking activity.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Imunoglobulina E/química , Imunoglobulina E/imunologia , Receptores de Antígenos de Linfócitos B/química , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Antialérgicos/química , Antialérgicos/metabolismo , Anticorpos Anti-Idiotípicos/química , Anticorpos Anti-Idiotípicos/metabolismo , Reagentes de Ligações Cruzadas , Humanos , Imunoglobulina E/biossíntese , Imunoglobulina E/metabolismo , Camundongos , Omalizumab/química , Omalizumab/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo
3.
Acta Neuropathol ; 127(6): 861-79, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24525765

RESUMO

Immunotherapeutic approaches are currently in the spotlight for their potential as disease-modifying treatments for neurodegenerative disorders. The discovery that α-synuclein (α-syn) can transmit from cell to cell in a prion-like fashion suggests that immunization might be a viable option for the treatment of synucleinopathies. This possibility has been bolstered by the development of next-generation active vaccination technology with short peptides-AFFITOPEs(®) (AFF)- that do not elicit an α-syn-specific T cell response. This approach allows for the production of long term, sustained, more specific, non-cross reacting antibodies suitable for the treatment of synucleinopathies, such as Parkinson's disease (PD). In this context, we screened a large library of peptides that mimic the C-terminus region of α-syn and discovered a novel set of AFF that identified α-syn oligomers. Next, the peptide that elicited the most specific response against α-syn (AFF 1) was selected for immunizing two different transgenic (tg) mouse models of PD and Dementia with Lewy bodies, the PDGF- and the mThy1-α-syn tg mice. Vaccination with AFF 1 resulted in high antibody titers in CSF and plasma, which crossed into the CNS and recognized α-syn aggregates. Active vaccination with AFF 1 resulted in decreased accumulation of α-syn oligomers in axons and synapses, accompanied by reduced degeneration of TH fibers in the caudo-putamen nucleus and by improvements in motor and memory deficits in both in vivo models. Clearance of α-syn involved activation of microglia and increased anti-inflammatory cytokine expression, further supporting the efficacy of this novel active vaccination approach for synucleinopathies.


Assuntos
Doença por Corpos de Lewy/terapia , Doença de Parkinson/terapia , Vacinação/métodos , Animais , Anticorpos/sangue , Anticorpos/líquido cefalorraquidiano , Axônios/patologia , Axônios/fisiologia , Núcleo Caudado/patologia , Núcleo Caudado/fisiopatologia , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos , Doença por Corpos de Lewy/patologia , Doença por Corpos de Lewy/fisiopatologia , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Transtornos da Memória/terapia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Microglia/fisiologia , Atividade Motora/fisiologia , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Degeneração Neural/terapia , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , Putamen/patologia , Putamen/fisiopatologia , Sinapses/patologia , Sinapses/fisiologia , Linfócitos T/patologia , Linfócitos T/fisiologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
FASEB J ; 27(3): 978-90, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23169771

RESUMO

Adhesion molecules play an important role in vascular biology because they mediate vascular stability, permeability, and leukocyte trafficking to and from tissues. Performing microarray analyses, we have recently identified activated leukocyte cell adhesion molecule (ALCAM) as an inflammation-induced gene in lymphatic endothelial cells (LECs). ALCAM belongs to the immunoglobulin superfamily and engages in homophilic as well as heterophilic interactions. In this study, we found ALCAM to be expressed at the protein level in human and murine lymphatic and blood vascular endothelial cells in vitro and in the vasculature of human and murine tissues in vivo. Functional in vitro experiments revealed that ALCAM mediates adhesive interactions, migration, and tube formation in LECs, suggesting a role for ALCAM in lymphatic vessel (LV) stability and in lymphangiogenesis. Furthermore, ALCAM supported dendritic cell (DC) adhesion to lymphatic endothelium. In agreement with these findings, experiments performed in ALCAM mice detected reduced LEC numbers in various tissues and defects in the formation of an organized LV network. Moreover, DC migration from lung to draining lymph nodes was compromised in ALCAM mice. Collectively, our data reveal a novel role for ALCAM in stabilizing LEC-LEC interactions and in the organization and function of the LV network.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Células Endoteliais/metabolismo , Proteínas Fetais/metabolismo , Linfonodos/metabolismo , Vasos Linfáticos/metabolismo , Animais , Antígenos CD/genética , Adesão Celular/fisiologia , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células Endoteliais/citologia , Proteínas Fetais/genética , Humanos , Pulmão/citologia , Pulmão/metabolismo , Linfonodos/citologia , Vasos Linfáticos/citologia , Camundongos , Camundongos Knockout
5.
Blood ; 120(11): 2249-58, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22855606

RESUMO

Dendritic cell (DC) migration via lymphatic vessels to draining lymph nodes (dLNs) is crucial for the initiation of adaptive immunity. We imaged this process by intravital microscopy (IVM) in the ear skin of transgenic mice bearing red-fluorescent vasculature and yellow-fluorescent DCs. DCs within lymphatic capillaries were rarely transported by flow, but actively migrated within lymphatics and were significantly faster than in the interstitium. Pharmacologic blockade of the Rho-associated protein kinase (ROCK), which mediates nuclear contraction and de-adhesion from integrin ligands, significantly reduced DC migration from skin to dLNs in steady-state. IVM revealed that ROCK blockade strongly reduced the velocity of interstitial DC migration, but only marginally affected intralymphatic DC migration. By contrast, during tissue inflammation, ROCK blockade profoundly decreased both interstitial and intralymphatic DC migration. Inhibition of intralymphatic migration was paralleled by a strong up-regulation of ICAM-1 in lymphatic endothelium, suggesting that during inflammation ROCK mediates de-adhesion of DC-expressed integrins from lymphatic-expressed ICAM-1. Flow chamber assays confirmed an involvement of lymphatic-expressed ICAM-1 and DC-expressed ROCK in DC crawling on lymphatic endothelium. Overall, our findings further define the role of ROCK in DC migration to dLNs and reveal a differential requirement for ROCK in intralymphatic DC crawling during steady-state and inflammation.


Assuntos
Movimento Celular , Células Dendríticas/metabolismo , Dermatite de Contato/metabolismo , Endotélio Linfático/imunologia , Quinases Associadas a rho/metabolismo , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Cruzamentos Genéticos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/patologia , Dermatite de Contato/tratamento farmacológico , Dermatite de Contato/imunologia , Dermatite de Contato/patologia , Endotélio Linfático/efeitos dos fármacos , Endotélio Linfático/metabolismo , Endotélio Linfático/patologia , Molécula 1 de Adesão Intercelular/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Microscopia de Vídeo , Inibidores de Proteínas Quinases/farmacologia , Quimera por Radiação , Proteínas Recombinantes de Fusão/metabolismo , Pele/efeitos dos fármacos , Pele/imunologia , Pele/metabolismo , Pele/patologia , Regulação para Cima/efeitos dos fármacos , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética
6.
Blood ; 118(1): 205-15, 2011 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-21596851

RESUMO

Chemokines and adhesion molecules up-regulated in lymphatic endothelial cells (LECs) during tissue inflammation are thought to enhance dendritic cell (DC) migration to draining lymph nodes, but the in vivo control of this process is not well understood. We performed a transcriptional profiling analysis of LECs isolated from murine skin and found that inflammation induced by a contact hypersensitivity (CHS) response up-regulated the adhesion molecules ICAM-1 and VCAM-1 and inflammatory chemokines. Importantly, the lymphatic markers Prox-1, VEGFR3, and LYVE-1 were significantly down-regulated during CHS. By contrast, skin inflammation induced by complete Freund adjuvant induced a different pattern of chemokine and lymphatic marker gene expression and almost no ICAM-1 up-regulation in LECs. Fluorescein isothiocyanate painting experiments revealed that DC migration to draining lymph nodes was more strongly increased in complete Freund adjuvant-induced than in CHS-induced inflammation. Surprisingly, DC migration did not correlate with the induction of CCL21 and ICAM-1 protein in LECs. Although the requirement for CCR7 signaling became further pronounced during inflammation, CCR7-independent signals had an additional, albeit moderate, impact on enhancing DC migration. Collectively, these findings indicate that DC migration in response to inflammation is stimulus-specific, mainly CCR7-dependent, and overall only moderately enhanced by LEC-induced genes other than CCL21.


Assuntos
Movimento Celular/imunologia , Células Dendríticas/imunologia , Dermatite de Contato/imunologia , Células Endoteliais/imunologia , Linfonodos/imunologia , Animais , Quimiocina CCL21/genética , Quimiocina CCL21/imunologia , Quimiocina CCL21/metabolismo , Células Dendríticas/citologia , Orelha Externa/imunologia , Feminino , Expressão Gênica/imunologia , Perfilação da Expressão Gênica , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Análise de Sequência com Séries de Oligonucleotídeos , Receptores CCR7/genética , Receptores CCR7/imunologia , Receptores CCR7/metabolismo , Regulação para Cima/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
Cancer Res ; 70(21): 8842-51, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20978206

RESUMO

Metastasis to regional lymph nodes (LN) is a prognostic indicator for cancer progression. There is a great demand for sensitive and noninvasive methods to detect metastasis to LNs. Whereas conventional in vivo imaging approaches have focused on the detection of cancer cells, lymphangiogenesis within tumor-draining LNs might be the earliest sign of metastasis. In mouse models of LN lymphangiogenesis, we found that systemically injected antibodies to lymphatic epitopes accumulated in the lymphatic vasculature in tissues and LNs. Using a (124)I-labeled antibody against the lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), we imaged, for the first time, inflammation- and tumor-draining LNs with expanded lymphatic networks in vivo by positron emission tomography (PET). Anti-LYVE-1 immuno-PET enabled visualization of lymphatic vessel expansion in LNs bearing metastases that were not detected by [(18)F]fluorodeoxyglucose-PET, which is clinically applied to detect cancer metastases. Immuno-PET with lymphatic-specific antibodies may open up new avenues for the early detection of metastasis, and the images obtained might be used as biomarkers for the progression of diseases associated with lymphangiogenesis.


Assuntos
Diagnóstico por Imagem , Glicoproteínas/imunologia , Inflamação/complicações , Linfonodos/diagnóstico por imagem , Linfangiogênese , Melanoma Experimental/complicações , Tomografia por Emissão de Pósitrons , Animais , Anticorpos Monoclonais/imunologia , Feminino , Fluordesoxiglucose F18 , Humanos , Inflamação/imunologia , Inflamação/patologia , Radioisótopos do Iodo/farmacocinética , Medições Luminescentes , Linfonodos/imunologia , Linfonodos/patologia , Metástase Linfática , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Prognóstico , Compostos Radiofarmacêuticos , Pele/metabolismo , Distribuição Tecidual , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/imunologia
8.
J Immunol ; 185(9): 5111-9, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20870948

RESUMO

The generation of Th17 cells has to be tightly controlled during an immune response. In this study, we report an increase in a CD44(high)CD62L(-) Th17 subset in mice deficient for the protein tyrosine kinase Tec. CD44(high)CD62L(-) Tec(-/-) CD4(+) T cells produced enhanced IL-17 upon activation, showed increased expression levels of IL-23R and RORγt, and IL-23-mediated expansion of Tec(-/-) CD4(+) T cells led to an increased production of IL-17. Tec(-/-) mice immunized with heat-killed Streptococcus pneumoniae displayed increased IL-17 expression levels in the lung postinfection with S. pneumoniae, and this correlated with enhanced pneumococcal clearance and reduced lung inflammation compared with Tec(+/+) mice. Moreover, naive Tec(-/-) OT-II CD4(+) T cells produced higher levels of IL-17 when cultured with OVA peptide-loaded bone marrow-derived dendritic cells that have been previously activated with heat-killed S. pneumoniae. Taken together, our data indicated a critical role for Tec in T cell-intrinsic signaling pathways that regulate the in vivo generation of CD44(high)CD62L(-) effector/memory Th17 populations.


Assuntos
Interleucina-17/imunologia , Proteínas Tirosina Quinases/imunologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem da Célula , Separação Celular , Citocinas/biossíntese , Encefalomielite Autoimune Experimental/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Receptores de Hialuronatos/imunologia , Receptores de Hialuronatos/metabolismo , Immunoblotting , Interleucina-17/metabolismo , Selectina L/imunologia , Selectina L/metabolismo , Camundongos , Camundongos Knockout , Pneumonia/imunologia , Proteínas Tirosina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/enzimologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/enzimologia
9.
Exp Cell Res ; 315(2): 336-47, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19007771

RESUMO

Lymphatic vessels play an important role in tissue fluid homeostasis, intestinal fat absorption and immunosurveillance. Furthermore, they are involved in pathologic conditions, such as tumor cell metastasis and chronic inflammation. In comparison to blood vessels, the molecular phenotype of lymphatic vessels is less well characterized. Performing comparative gene expression analysis we have recently found that coxsackie- and adenovirus receptor (CAR) is significantly more highly expressed in cultured human, skin-derived lymphatic endothelial cells (LECs), as compared to blood vascular endothelial cells. Here, we have confirmed these results at the protein level, using Western blot and FACS analysis. Immunofluorescence performed on human skin confirmed that CAR is expressed at detectable levels in lymphatic vessels, but not in blood vessels. To address the functional significance of CAR expression, we modulated CAR expression levels in cultured LECs in vitro by siRNA- and vector-based transfection approaches. Functional assays performed with the transfected cells revealed that CAR is involved in distinct cellular processes in LECs, such as cell adhesion, migration, tube formation and the control of vascular permeability. In contrast, no effect of CAR on LEC proliferation was observed. Overall, our data suggest that CAR stabilizes LEC-LEC interactions in the skin and may contribute to lymphatic vessel integrity.


Assuntos
Células Endoteliais/metabolismo , Vasos Linfáticos/metabolismo , Receptores Virais/fisiologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Dextranos/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Prepúcio do Pênis/citologia , Prepúcio do Pênis/efeitos dos fármacos , Prepúcio do Pênis/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Recém-Nascido , Molécula 1 de Adesão Intercelular/genética , Queratinócitos/metabolismo , Vasos Linfáticos/citologia , Masculino , Glicoproteínas de Membrana/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Interferente Pequeno/genética , Receptores Virais/genética , Receptores Virais/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Proteínas de Transporte Vesicular/genética
10.
Blood ; 110(9): 3158-67, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17625067

RESUMO

Lymphangiogenesis is involved in tumor cell metastasis and plays a major role in chronic inflammatory disorders. To investigate the role of lymphangiogenesis in inflammation, we induced and maintained delayed-type hypersensitivity (DTH) reactions in the ears of mice and then analyzed the resulting lymphangiogenesis in the inflamed tissue and draining lymph nodes (LNs) by quantitative fluorescence-activated cell sorting (FACS) and by immunofluorescence. Long-lasting inflammation induced a significant increase in the number of lymphatic endothelial cells, not only in the inflamed ears but also in the ear-draining auricular LNs. Inflammation-induced lymphangiogenesis was potently blocked by systemic administration of a vascular endothelial growth factor (VEGF)-A neutralizing antibody. Surprisingly, tissue inflammation specifically induced LN lymphangiogenesis but not LN angiogenesis. These findings were explained by analysis of both VEGF-A protein and mRNA levels, which revealed that VEGF-A was expressed at high mRNA and protein levels in inflamed ears but that expression was increased only at the protein level in activated LNs. Inflammation-induced lymphangiogenesis in LNs was independent of the presence of nodal B lymphocytes, as shown in B cell-deficient mice. Our data reveal that chronic inflammation actively induces lymphangiogenesis in LNs, which is controlled remotely, by lymphangiogenic factors produced at the site of inflammation.


Assuntos
Inflamação/metabolismo , Linfonodos , Linfangiogênese/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Doença Crônica , Dermatite Alérgica de Contato/patologia , Células Endoteliais/patologia , Feminino , Inflamação/genética , Inflamação/patologia , Linfonodos/metabolismo , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Pele/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...