Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 8(329): 329ra34, 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26962158

RESUMO

The vast majority of ovarian cancer-related deaths are caused by metastatic dissemination of tumor cells, resulting in subsequent organ failure. However, despite our increased understanding of the physiological processes involved in tumor metastasis, there are no clinically approved drugs that have made a major impact in increasing the overall survival of patients with advanced, metastatic ovarian cancer. We identified prosaposin (psap) as a potent inhibitor of tumor metastasis, which acts via stimulation of p53 and the antitumorigenic protein thrombospondin-1 (TSP-1) in bone marrow-derived cells that are recruited to metastatic sites. We report that more than 97% of human serous ovarian tumors tested express CD36, the receptor that mediates the proapoptotic activity of TSP-1. Accordingly, we sought to determine whether a peptide derived from psap would be effective in treating this form of ovarian cancer. To that end, we developed a cyclic peptide with drug-like properties derived from the active sequence in psap. The cyclic psap peptide promoted tumor regression in a patient-derived tumor xenograft model of metastatic ovarian cancer. Thus, we hypothesize that a therapeutic agent based on this psap peptide would have efficacy in treating patients with metastatic ovarian cancer.


Assuntos
Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Peptídeos Cíclicos/uso terapêutico , Saposinas/química , Trombospondina 1/farmacologia , Microambiente Tumoral , Aminoácidos/metabolismo , Animais , Antígenos CD36/metabolismo , Carcinoma Epitelial do Ovário , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclização , Modelos Animais de Doenças , Feminino , Humanos , Camundongos Endogâmicos C57BL , Gradação de Tumores , Neoplasias Epiteliais e Glandulares/secundário , Neoplasias Ovarianas/secundário , Peptídeos Cíclicos/farmacologia , Análise de Regressão , Análise Serial de Tecidos , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Stem Cell Res Ther ; 5(6): 143, 2014 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-25528303

RESUMO

INTRODUCTION: Inflammation plays a crucial role in tissue regeneration, wound healing, and the success of tissue-engineered constructs. The aim of this study was to investigate the influence of human umbilical vein endothelial cells (ECs) on leukocyte transmigration when co-cultured with primary human bone marrow-derived multipotent stromal cells (MSCs). METHODS: MSCs with and without ECs were cultured in poly (L-lactide-co-1, 5-dioxepan-2-one) (poly (LLA-co-DXO)) scaffolds for 1 week in vitro in a bioreactor system, after which they were implanted subcutaneously in non-obese diabetic/severe combined immunodeficient mice. After 1 and 3 weeks, scaffolds were retrieved, and the mRNA expression of interleukin 1-beta (IL-1ß), IL-6, IL-10, hypoxia-inducible factor 1-alpha (HIF-1α), HIF-1ß, and mammalian target of rapamycin was examined by real-time reverse transcription-polymerase chain reaction. Furthermore, immunofluorescent staining was performed for IL-1ß, IL-6, neutrophils, and CD11b. In addition, Western blotting was done for IL-1ß and IL-6. Leukocyte transmigration genes and genes in Toll-like receptor pathways, expressed by MSCs cultured in vitro with or without ECs, were further investigated with a microarray dataset. RESULTS: In vitro, genes involved in leukocyte transmigration and Toll-like receptor pathways were clearly influenced by the addition of ECs. Platelet/endothelial cell adhesion molecule-1 (PECAM-1) and cadherin-5 (CDH5), both genes involved in leukocyte transmigration, were expressed significantly higher in the MSC/EC group. CONCLUSIONS: The recruitment of leukocytes into tissue-engineered constructs with MSCs is strongly influenced by the addition of ECs via activation of leukocyte transmigration and Toll-like receptor pathways.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Leucócitos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptores Toll-Like/metabolismo , Migração Transendotelial e Transepitelial , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Caderinas/genética , Caderinas/metabolismo , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/fisiologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Engenharia Tecidual , Alicerces Teciduais , Receptores Toll-Like/genética
3.
Tissue Eng Part A ; 20(19-20): 2590-603, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24957363

RESUMO

Akt kinase is a central signal transduction node that integrates extracellular cues that regulate cell migratory, proliferative, and morphological functions during angiogenesis. However, how Akt activity is modulated and contributes to subsequent vessel maturation is unclear. In this study we investigated the role of Akt1 in vessel maturation using human dermal microvascular endothelial cells (HDMVECs) expressing constitutively active and hemiphosphorylated Akt1 epi-alleles with graded kinase activity. HDMVECs expressing Akt1 epi-alleles were analyzed in vivo in a tissue engineering setting using a model of angiogenesis comprising cell-seeded poly-L-lactic acid scaffolds implanted subcutaneously into NOD/SCID murine hosts. The resultant intraimplant microvasculature was quantified for vascular parameters, including vessel diameter, perfusion, vascular density, and pericyte coverage. We found that constitutive Akt1 kinase activity in implanted HDMVECs correlated with loss of neovasculature function. Further, we found that the presence of coimplanted vascular smooth muscle cells (vSMCs) in the implants failed to promote blood vessel growth and maturation in a graded, Akt1 kinase activity-dependent manner. These results indicate that constitutive Akt1 activity disrupts the normal blood vessel growth and maturation. Therefore, we suggest that a downregulation of Akt1 activity is necessary for vSMC-induced maturation of newly formed blood vessels to occur.


Assuntos
Células Endoteliais/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Engenharia Tecidual , Animais , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/transplante , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais
4.
Stem Cell Res Ther ; 5(1): 23, 2014 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-24533904

RESUMO

INTRODUCTION: Rapid establishment of functional blood vessels is a prerequisite for successful tissue engineering. During vascular development, endothelial cells (ECs) and perivascular cells assemble into a complex regulating proliferation of ECs, vessel diameter and production of extracellular matrix proteins. The aim of this study was to evaluate the ability of mesenchymal stem cells (MSCs) to establish an endothelial-perivascular complex in tissue-engineered constructs comprising ECs and MSCs. METHODS: Primary human ECs and MSCs were seeded onto poly(L-lactide-co-1,5-dioxepan-2-one) (poly(LLA-co-DXO)) scaffolds and grown in dynamic culture before subcutaneous implantation in immunocompromised mice for 1 and 3 weeks. Cellular activity, angiogenic stimulation and vascular assembly in cell/scaffold constructs seeded with ECs or ECs/MSCs in a 5:1 ratio was monitored with real-time RT-PCR, ELISA and immunohistochemical microscopy analysis. RESULTS: A quiescent phenotype of ECs was generated, by adding MSCs to the culture system. Decreased proliferation of ECs, in addition to up-regulation of selected markers for vascular maturation was demonstrated. Baseline expression of VEGFa was higher for MSCs compared with EC (P<0.001), with subsequent up-regulated VEGFa-expression for EC/MSC constructs before (P<0.05) and after implantation (P<0.01). Furthermore, an inflammatory response with CD11b+cells was generated from implantation of human cells. At the end of the 3 week experimental period, a higher vascular density was shown for both cellular constructs compared with empty control scaffolds (P<0.01), with the highest density of capillaries being generated in constructs comprising both ECs and MSCs. CONCLUSIONS: Induction of a quiescent phenotype of ECs associated with vascular maturation can be achieved by co-seeding with MSCs. Hence, MSCs can be appropriate perivascular cells for tissue-engineered constructs.


Assuntos
Diferenciação Celular , Células Endoteliais da Veia Umbilical Humana/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica , Animais , Proliferação de Células , Células Cultivadas , Fator 1 de Crescimento de Fibroblastos/genética , Fator 1 de Crescimento de Fibroblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Engenharia Tecidual , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Mol Cancer Res ; 12(5): 754-64, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24574516

RESUMO

UNLABELLED: The angiogenic switch, a rate-limiting step in tumor progression, has already occurred by the time most human tumors are detectable. However, despite significant study of the mechanisms controlling this switch, the kinetics and reversibility of the process have not been explored. The stability of the angiogenic phenotype was examined using an established human liposarcoma xenograft model. Nonangiogenic cells inoculated into immunocompromised mice formed microscopic tumors that remained dormant for approximately 125 days (vs. <40 days for angiogenic cells) whereupon the vast majority (>95%) initiated angiogenic growth with second-order kinetics. These original, clonally derived angiogenic tumor cells were passaged through four in vivo cycles. At each cycle, a new set of single-cell clones was established from the most angiogenic clone and characterized for in vivo for tumorigenic activity. A total of 132 single-cell clones were tested in the second, third, and fourth in vivo passage. Strikingly, at each passage, a portion of the single-cell clones formed microscopic, dormant tumors. Following dormancy, like the original cell line, these revertant tumors spontaneously switched to the angiogenic phenotype. Finally, revertant clones were transcriptionally profiled and their angiogenic output determined. Collectively, these data demonstrate that the angiogenic phenotype in tumors is malleable and can spontaneously revert to the nonangiogenic phenotype in a population of human tumor cells. IMPLICATIONS: Leveraging the rate of reversion to the nonangiogenic phenotype and tumor dormancy may be a novel anticancer strategy.


Assuntos
Lipossarcoma/irrigação sanguínea , Lipossarcoma/patologia , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Expressão Gênica , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos SCID , Neovascularização Patológica/patologia , Fenótipo
6.
Stem Cell Res Ther ; 4(3): 52, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23683577

RESUMO

INTRODUCTION: A major determinant of the potential size of cell/scaffold constructs in tissue engineering is vascularization. The aims of this study were twofold: first to determine the in vitro angiogenic and osteogenic gene-expression profiles of endothelial cells (ECs) and mesenchymal stem cells (MSCs) cocultured in a dynamic 3D environment; and second, to assess differentiation and the potential for osteogenesis after in vivo implantation. METHODS: MSCs and ECs were grown in dynamic culture in poly(L-lactide-co-1,5-dioxepan-2-one) (poly(LLA-co-DXO)) copolymer scaffolds for 1 week, to generate three-dimensional endothelial microvascular networks. The constructs were then implanted in vivo, in a murine model for ectopic bone formation. Expression of selected genes for angiogenesis and osteogenesis was studied after a 1-week culture in vitro. Human cell proliferation was assessed as expression of ki67, whereas α-smooth muscle actin was used to determine the perivascular differentiation of MSCs. Osteogenesis was evaluated in vivo through detection of selected markers, by using real-time RT-PCR, alkaline phosphatase (ALP), Alizarin Red, hematoxylin/eosin (HE), and Masson trichrome staining. RESULTS: The results show that endothelial microvascular networks could be generated in a poly(LLA-co-DXO) scaffold in vitro and sustained after in vivo implantation. The addition of ECs to MSCs influenced both angiogenic and osteogenic gene-expression profiles. Furthermore, human ki67 was upregulated before and after implantation. MSCs could support functional blood vessels as perivascular cells independent of implanted ECs. In addition, the expression of ALP was upregulated in the presence of endothelial microvascular networks. CONCLUSIONS: This study demonstrates that copolymer poly(LLA-co-DXO) scaffolds can be prevascularized with ECs and MSCs. Although a local osteoinductive environment is required to achieve ectopic bone formation, seeding of MSCs with or without ECs increases the osteogenic potential of tissue-engineered constructs.


Assuntos
Microvasos/citologia , Engenharia Tecidual , Alicerces Teciduais , Actinas/metabolismo , Animais , Técnicas de Cultura de Células/instrumentação , Diferenciação Celular , Técnicas de Cocultura , Células Endoteliais da Veia Umbilical Humana , Humanos , Antígeno Ki-67/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microvasos/metabolismo , Osteogênese , Poliésteres/química , Transcriptoma , Transplante Heterólogo
7.
J Tissue Eng ; 3(1): 2041731412443236, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22511994

RESUMO

To optimize culture conditions for in vitro prevascularization of tissue-engineered bone constructs, the development of organotypic blood vessels under osteogenic stimulatory conditions (OM) was investigated. Coculture of endothelial cells and mesenchymal stem cells was used to assess proangiogenic effects of mesenchymal stem cells on endothelial cells. Four different culture conditions were evaluated for their effect on development of microvascular endothelial cell networks. Mineralization, deposition of extracellular matrix, and perivascular gene expression were studied in OM. After 3 days, endothelial cells established elongated capillary-like networks, and upregulated expression of vascular markers was seen. After 15 days, all parameters evaluated were significantly increased for cultures in OM. Mature networks developed in OM presented lumens enveloped by basement membrane-like collagen IV, with obvious mineralization and upregulated perivascular gene expression from mesenchymal stem cells. Our results suggest osteogenic stimulatory conditions to be appropriate for in vitro development of vascularized bone implants for tissue engineering.

8.
J Tissue Eng Regen Med ; 5(4): e52-62, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20865694

RESUMO

The success of tissue engineering depends on the rapid and efficient formation of a functional blood vasculature. Adult blood vessels comprise endothelial cells and perivascular mural cells that assemble into patent tubules ensheathed by a basement membrane during angiogenesis. Using individual vessel components, we characterized intra-scaffold microvessel self-assembly efficiency in a physiological in vivo tissue engineering implant context. Primary human microvascular endothelial and vascular smooth muscle cells were seeded at different ratios in poly-L-lactic acid (PLLA) scaffolds enriched with basement membrane proteins (Matrigel) and implanted subcutaneously into immunocompromised mice. Temporal intra-scaffold microvessel formation, anastomosis and perfusion were monitored by immunohistochemical, flow cytometric and in vivo multiphoton fluorescence microscopy analysis. Vascularization in the tissue-engineering context was strongly enhanced in implants seeded with a complete complement of blood vessel components: human microvascular endothelial and vascular smooth muscle cells in vivo assembled a patent microvasculature within Matrigel-enriched PLLA scaffolds that anastomosed with the host circulation during the first week of implantation. Multiphoton fluorescence angiographic analysis of the intra-scaffold microcirculation showed a uniform, branched microvascular network. 3D image reconstruction analysis of human pulmonary artery smooth muscle cell (hPASMC) distribution within vascularized implants was non-random and displayed a preferential perivascular localization. Hence, efficient microvessel self-assembly, anastomosis and establishment of a functional microvasculture in the native hypoxic in vivo tissue engineering context is promoted by providing a complete set of vascular components.


Assuntos
Neovascularização Fisiológica , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Adesão Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Angiofluoresceinografia , Humanos , Imuno-Histoquímica , Ácido Láctico/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Microcirculação/efeitos dos fármacos , Microvasos/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Poliésteres , Polímeros/farmacologia , Artéria Pulmonar/citologia
9.
PLoS One ; 4(6): e5798, 2009 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-19495422

RESUMO

BACKGROUND: Blood vessels comprise endothelial cells, mural cells (pericytes/vascular smooth muscle cells) and basement membrane. During angiogenesis, mural cells are recruited to sprouting endothelial cells and define a stabilizing context, comprising cell-cell contacts, secreted growth factors and extracellular matrix components, that drives vessel maturation and resistance to anti-angiogenic therapeutics. METHODS AND FINDINGS: To better understand the basis for mural cell regulation of angiogenesis, we conducted high content imaging analysis on a microtiter plate format in vitro organotypic blood vessel system comprising primary human endothelial cells co-cultured with primary human mural cells. We show that endothelial cells co-cultured with mural cells undergo an extensive series of phenotypic changes reflective of several facets of blood vessel formation and maturation: Loss of cell proliferation, pathfinding-like cell migration, branching morphogenesis, basement membrane extracellular matrix protein deposition, lumen formation, anastamosis and development of a stabilized capillary-like network. This phenotypic sequence required endothelial-mural cell-cell contact, mural cell-derived VEGF and endothelial VEGFR2 signaling. Inhibiting formation of adherens junctions or basement membrane structures abrogated network formation. Notably, inhibition of mural cell VEGF expression could not be rescued by exogenous VEGF. CONCLUSIONS: These results suggest a unique role for mural cell-associated VEGF in driving vessel formation and maturation.


Assuntos
Vasos Sanguíneos/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Junções Aderentes/metabolismo , Inibidores da Angiogênese/farmacologia , Membrana Basal/metabolismo , Capilares/metabolismo , Comunicação Celular , Proliferação de Células , Técnicas de Cocultura , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Fisiológica , Interferência de RNA
10.
Regul Pept ; 135(1-2): 78-84, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16725215

RESUMO

The proinflammatory agent tumour necrosis factor alpha (TNFalpha) is one of several agents causing vascular leakage. The N-terminal domain of CgA, vasostatin-I (CgA1-76), has recently been reported to inhibit TNFalpha induced gap formation in human umbilical venous endothelial cells. Here we report on the effect of recombinant human CgA1-78, vasostatin-I, on TNFalpha induced gap formation in two model systems of vascular leakage in arterial endothelial cells of bovine pulmonary (BPAEC) and coronary (BCAEC) origin. Vasostatin-I inhibited the TNFalpha induced gap formation in both models, being inactive in the unstimulated cells. The phosphorylation of p38MAP kinase in TNFalpha activated BPAEC was markedly attenuated in the presence of vasostatin-I and the inhibitory effect corresponded to that of the specific p38MAPK inhibitor SB203580. Vasostatin-I also inhibited the phosphorylation of p38MAPK induced by both thrombin and pertussis toxin in these cells. The results demonstrate that vasostatin-I has inhibitory effects on TNFalpha-induced disruption of confluent layers of cultured pulmonary and coronary arterial endothelial cells. This suggests that vasostatin-I may affect endothelial barrier dysfunction also in arterial vascular beds. Furthermore, the inhibitory activity of vasostatin-I may be associated with the p38MAPK signalling cascade via a pertussis toxin sensitive, presumably Galphai coupled mechanism.


Assuntos
Cromogranina A/metabolismo , Vasos Coronários/citologia , Células Endoteliais/metabolismo , Fragmentos de Peptídeos/metabolismo , Artéria Pulmonar/citologia , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Bovinos , Células Cultivadas , Cromogranina A/genética , Células Endoteliais/citologia , Inibidores Enzimáticos/metabolismo , Humanos , Imidazóis/metabolismo , Fragmentos de Peptídeos/genética , Toxina Pertussis/metabolismo , Piridinas/metabolismo , Trombina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Regul Pept ; 134(1): 30-7, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16445995

RESUMO

Vasostatin-I (CgA1-76) is a naturally occurring and biologically active N-terminal peptide derived from chromogranin A (CgA), produced and secreted at high concentrations by neuroendocrine tissues and also from a range of neuroendocrine tumors. This study aims to examine the hypothesis that in the absence of classical protein receptors CgA1-76 may, like its two derived peptides CgA1-40 and CgA47-66, perturb the lipid microenvironment of other membrane receptors, as a basis for the largely inhibitory activities of these CgA peptides. The nature of the interactions between phospholipids and vasostatin-derived fragments was studied in the Langmuir film balance apparatus at 37 degrees C. The synthetic peptides CgA1-40 and CgA47-66 and a recombinant fragment (VS-I) containing vasostatin-I (Ser-Thr-Ala-CgA1-78) were compared for their effects on monolayers of phosphatidylcholine and phosphatidylethanolamine from pig brain and defined species of phosphatidylserine. Marked differences in surface pressure-area isotherms and phase-transition plateaus were apparent with the three classes of phospholipids on VS-I, CgA1-40 and CgA47-66 in physiological buffer or pure water. The results indicate that VS-I and CgA47-66 at 5-10 nM concentrations may engage in electrostatic as well as hydrophobic interactions with membrane-relevant phospholipids at physiological conditions, VS-I in particular enhancing the fluidity of saturated species of phosphatidylserine.


Assuntos
Cromograninas/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Animais , Encéfalo/metabolismo , Cromogranina A , Cromograninas/farmacologia , Interações Medicamentosas , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Fragmentos de Peptídeos/farmacologia , Fosfolipídeos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/fisiologia , Suínos , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...