Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Commun Biol ; 7(1): 618, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783087

RESUMO

Endothelial cells (ECs) are highly glycolytic, but whether they generate glycolytic intermediates via gluconeogenesis (GNG) in glucose-deprived conditions remains unknown. Here, we report that glucose-deprived ECs upregulate the GNG enzyme PCK2 and rely on a PCK2-dependent truncated GNG, whereby lactate and glutamine are used for the synthesis of lower glycolytic intermediates that enter the serine and glycerophospholipid biosynthesis pathways, which can play key roles in redox homeostasis and phospholipid synthesis, respectively. Unexpectedly, however, even in normal glucose conditions, and independent of its enzymatic activity, PCK2 silencing perturbs proteostasis, beyond its traditional GNG role. Indeed, PCK2-silenced ECs have an impaired unfolded protein response, leading to accumulation of misfolded proteins, which due to defective proteasomes and impaired autophagy, results in the accumulation of protein aggregates in lysosomes and EC demise. Ultimately, loss of PCK2 in ECs impaired vessel sprouting. This study identifies a role for PCK2 in proteostasis beyond GNG.


Assuntos
Células Endoteliais , Gluconeogênese , Fosfoenolpiruvato Carboxiquinase (GTP) , Proteostase , Gluconeogênese/genética , Humanos , Células Endoteliais/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo , Glucose/metabolismo , Autofagia , Resposta a Proteínas não Dobradas , Fosfoenolpiruvato Carboxiquinase (ATP)
2.
MethodsX ; 11: 102446, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37928105

RESUMO

Angiogenesis and lymphangiogenesis, the formation of new blood or lymphatic vessels, respectively, from preexisting vasculature is essential during embryonic development, but also occurs during tissue repair and in pathological conditions (cancer; ocular disease; ischemic, infectious and inflammatory disorders), which are all characterized to a certain extent by inflammatory conditions. Hence, a rapid, inexpensive, feasible / technically easy, reliable assay of inflammation-induced (lymph-)angiogenesis is highly valuable. In this context, the corneal thermal cauterization assay in mice is a simple, low-cost, reproducible, insightful and labor-saving assay to gauge the role of inflammation in angiogenesis and lymphangiogenesis. However, to the best of our knowledge, there is no standardized protocol to perform this assay. Here, we provide a step-by-step description of the model's procedures, which include:•The thermal cauterization of the corneas,•Enucleation and dissection of the corneas,•Subsequent immunofluorescence staining of the neovasculature, and morphometric analysis. We also discuss ethical considerations and aspects related to animal welfare guidelines. Altogether, this paper will help to increase the reproducibility of the corneal thermal cauterization model and facilitate its use for angiogenesis and lymphangiogenesis research.

3.
Commun Biol ; 6(1): 648, 2023 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-37330599

RESUMO

Translation of academic results into clinical practice is a formidable unmet medical need. Single-cell RNA-sequencing (scRNA-seq) studies generate long descriptive ranks of markers with predicted biological function, but without functional validation, it remains challenging to know which markers truly exert the putative function. Given the lengthy/costly nature of validation studies, gene prioritization is required to select candidates. We address these issues by studying tip endothelial cell (EC) marker genes because of their importance for angiogenesis. Here, by tailoring Guidelines On Target Assessment for Innovative Therapeutics, we in silico prioritize previously unreported/poorly described, high-ranking tip EC markers. Notably, functional validation reveals that four of six candidates behave as tip EC genes. We even discover a tip EC function for a gene lacking in-depth functional annotation. Thus, validating prioritized genes from scRNA-seq studies offers opportunities for identifying targets to be considered for possible translation, but not all top-ranked scRNA-seq markers exert the predicted function.


Assuntos
Perfilação da Expressão Gênica , Transcriptoma , Perfilação da Expressão Gênica/métodos
4.
STAR Protoc ; 3(4): 101691, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36173713

RESUMO

Tumor vessel co-option, a process in which cancer cells "hijack" pre-existing blood vessels to grow and invade healthy tissue, is poorly understood but is a proposed resistance mechanism against anti-angiogenic therapy (AAT). Here, we describe protocols for establishing murine renal (RENCA) and breast (4T1) cancer lung vessel co-option metastases models. Moreover, we outline a reproducible protocol for single-cell isolation from murine lung metastases using magnetic-activated cell sorting as well as immunohistochemical stainings to distinguish vessel co-option from angiogenesis. For complete details on the use and execution of this protocol, please refer to Teuwen et al. (2021).


Assuntos
Neoplasias Pulmonares , Neovascularização Patológica , Camundongos , Animais , Neovascularização Patológica/patologia , Células Endoteliais , Neoplasias Pulmonares/patologia , Modelos Animais de Doenças
5.
Circ Res ; 127(4): 466-482, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32404031

RESUMO

RATIONALE: Endothelial cells (ECs) are highly glycolytic and generate the majority of their energy via the breakdown of glucose to lactate. At the same time, a main role of ECs is to allow the transport of glucose to the surrounding tissues. GLUT1 (glucose transporter isoform 1/Slc2a1) is highly expressed in ECs of the central nervous system (CNS) and is often implicated in blood-brain barrier (BBB) dysfunction, but whether and how GLUT1 controls EC metabolism and function is poorly understood. OBJECTIVE: We evaluated the role of GLUT1 in endothelial metabolism and function during postnatal CNS development as well as at the adult BBB. METHODS AND RESULTS: Inhibition of GLUT1 decreases EC glucose uptake and glycolysis, leading to energy depletion and the activation of the cellular energy sensor AMPK (AMP-activated protein kinase), and decreases EC proliferation without affecting migration. Deletion of GLUT1 from the developing postnatal retinal endothelium reduces retinal EC proliferation and lowers vascular outgrowth, without affecting the number of tip cells. In contrast, in the brain, we observed a lower number of tip cells in addition to reduced brain EC proliferation, indicating that within the CNS, organotypic differences in EC metabolism exist. Interestingly, when ECs become quiescent, endothelial glycolysis is repressed, and GLUT1 expression increases in a Notch-dependent fashion. GLUT1 deletion from quiescent adult ECs leads to severe seizures, accompanied by neuronal loss and CNS inflammation. Strikingly, this does not coincide with BBB leakiness, altered expression of genes crucial for BBB barrier functioning nor reduced vascular function. Instead, we found a selective activation of inflammatory and extracellular matrix related gene sets. CONCLUSIONS: GLUT1 is the main glucose transporter in ECs and becomes uncoupled from glycolysis during quiescence in a Notch-dependent manner. It is crucial for developmental CNS angiogenesis and adult CNS homeostasis but does not affect BBB barrier function.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/irrigação sanguínea , Células Endoteliais/metabolismo , Transportador de Glucose Tipo 1/fisiologia , Neovascularização Fisiológica , Vasos Retinianos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Encéfalo/citologia , Movimento Celular , Proliferação de Células , Células Endoteliais/fisiologia , Endotélio , Endotélio Vascular/fisiologia , Metabolismo Energético , Glucose/metabolismo , Transportador de Glucose Tipo 1/antagonistas & inibidores , Glicólise , Humanos , Camundongos , Retina/citologia
6.
Nat Metab ; 1(7): 666-675, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-32694649

RESUMO

Lymphatic vessels (LVs), lined by lymphatic endothelial cells (LECs), are indispensable for life1. However, the role of metabolism in LECs has been incompletely elucidated. In the present study, it is reported that LEC-specific loss of OXCT1, a key enzyme of ketone body oxidation2, reduces LEC proliferation, migration and vessel sprouting in vitro and impairs lymphangiogenesis in development and disease in Prox1ΔOXCT1 mice. Mechanistically, OXCT1 silencing lowers acetyl-CoA levels, tricarboxylic acid cycle metabolite pools, and nucleotide precursor and deoxynucleotide triphosphate levels required for LEC proliferation. Ketone body supplementation to LECs induces the opposite effects. Notably, elevation of lymph ketone body levels by a high-fat, low-carbohydrate ketogenic diet or by administration of the ketone body ß-hydroxybutyrate increases lymphangiogenesis after corneal injury and myocardial infarction. Intriguingly, in a mouse model of microsurgical ablation of LVs in the tail, which repeats features of acquired lymphoedema in humans, the ketogenic diet improves LV function and growth, reduces infiltration of anti-lymphangiogenic immune cells and decreases oedema, suggesting a novel dietary therapeutic opportunity.


Assuntos
Dieta , Corpos Cetônicos/metabolismo , Vasos Linfáticos/metabolismo , Animais , Dieta Cetogênica , Humanos , Camundongos , Oxirredução
7.
Angiogenesis ; 20(4): 599-613, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28875379

RESUMO

Blockade of the glycolytic activator PFKFB3 in cancer cells (using a maximum tolerable dose of 70 mg/kg of the PFKFB3 blocker 3PO) inhibits tumor growth in preclinical models and is currently being tested as a novel anticancer treatment in phase I clinical trials. However, a detailed preclinical analysis of the effects of such maximum tolerable dose of a PFKFB3 blocker on the tumor vasculature is lacking, even though tumor endothelial cells are hyper-glycolytic. We report here that a high dose of 3PO (70 mg/kg), which inhibits cancer cell proliferation and reduces primary tumor growth, causes tumor vessel disintegration, suppresses endothelial cell growth for protracted periods, (model-dependently) aggravates tumor hypoxia, and compromises vascular barrier integrity, thereby rendering tumor vessels more leaky and facilitating cancer cell intravasation and dissemination. These findings contrast to the effects of a low dose of 3PO (25 mg/kg), which induces tumor vessel normalization, characterized by vascular barrier tightening and maturation, but reduces cancer cell intravasation and metastasis. Our findings highlight the importance of adequately dosing a glycolytic inhibitor for anticancer treatment.


Assuntos
Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Fosfofrutoquinase-2/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Melanoma Experimental/ultraestrutura , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neoplasias/patologia , Neovascularização Patológica/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Fosfofrutoquinase-2/metabolismo , Piridinas/farmacologia
8.
Cell Rep ; 18(12): 3005-3017, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28329691

RESUMO

Peripheral tolerance is crucial for avoiding activation of self-reactive T cells to tissue-restricted antigens. Sterile tissue injury can break peripheral tolerance, but it is unclear how autoreactive T cells get activated in response to self. An example of a sterile injury is myocardial infarction (MI). We hypothesized that tissue necrosis is an activator of dendritic cells (DCs), which control tolerance to self-antigens. DC subsets of a murine healthy heart consisted of IRF8-dependent conventional (c)DC1, IRF4-dependent cDC2, and monocyte-derived DCs. In steady state, cardiac self-antigen α-myosin was presented in the heart-draining mediastinal lymph node (mLN) by cDC1s, driving the proliferation of antigen-specific CD4+ TCR-M T cells and their differentiation into regulatory cells (Tregs). Following MI, all DC subsets infiltrated the heart, whereas only cDCs migrated to the mLN. Here, cDC2s induced TCR-M proliferation and differentiation into interleukin-(IL)-17/interferon-(IFN)γ-producing effector cells. Thus, cardiac-specific autoreactive T cells get activated by mature DCs following myocardial infarction.


Assuntos
Células Dendríticas/imunologia , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Linfócitos T/imunologia , Animais , Antígeno CD11c/metabolismo , Linfócitos T CD4-Positivos/imunologia , Movimento Celular , Fatores Reguladores de Interferon/metabolismo , Linfonodos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/patologia , Miocárdio/patologia , Miosinas/metabolismo , Fenótipo , Fatores de Transcrição/metabolismo
9.
Cell Stem Cell ; 20(3): 360-373.e7, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-27889318

RESUMO

Whether new neurons are added in the postnatal cerebral cortex is still debated. Here, we report that the meninges of perinatal mice contain a population of neurogenic progenitors formed during embryonic development that migrate to the caudal cortex and differentiate into Satb2+ neurons in cortical layers II-IV. The resulting neurons are electrically functional and integrated into local microcircuits. Single-cell RNA sequencing identified meningeal cells with distinct transcriptome signatures characteristic of (1) neurogenic radial glia-like cells (resembling neural stem cells in the SVZ), (2) neuronal cells, and (3) a cell type with an intermediate phenotype, possibly representing radial glia-like meningeal cells differentiating to neuronal cells. Thus, we have identified a pool of embryonically derived radial glia-like cells present in the meninges that migrate and differentiate into functional neurons in the neonatal cerebral cortex.


Assuntos
Diferenciação Celular , Movimento Celular , Córtex Cerebral/citologia , Meninges/citologia , Neurogênese , Neuroglia/citologia , Neurônios/citologia , Animais , Animais Recém-Nascidos , Linhagem da Célula , Embrião de Mamíferos/citologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Nestina/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Reprodutibilidade dos Testes , Análise de Célula Única , Esferoides Celulares/citologia , Coloração e Rotulagem , Transcriptoma/genética
10.
Nat Commun ; 7: 12240, 2016 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-27436424

RESUMO

During vessel sprouting, endothelial cells (ECs) dynamically rearrange positions in the sprout to compete for the tip position. We recently identified a key role for the glycolytic activator PFKFB3 in vessel sprouting by regulating cytoskeleton remodelling, migration and tip cell competitiveness. It is, however, unknown how glycolysis regulates EC rearrangement during vessel sprouting. Here we report that computational simulations, validated by experimentation, predict that glycolytic production of ATP drives EC rearrangement by promoting filopodia formation and reducing intercellular adhesion. Notably, the simulations correctly predicted that blocking PFKFB3 normalizes the disturbed EC rearrangement in high VEGF conditions, as occurs during pathological angiogenesis. This interdisciplinary study integrates EC metabolism in vessel sprouting, yielding mechanistic insight in the control of vessel sprouting by glycolysis, and suggesting anti-glycolytic therapy for vessel normalization in cancer and non-malignant diseases.


Assuntos
Glicólise , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica , Trifosfato de Adenosina/metabolismo , Antígenos CD/metabolismo , Caderinas/antagonistas & inibidores , Caderinas/metabolismo , Simulação por Computador , Técnicas de Silenciamento de Genes , Glicólise/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Indóis/farmacologia , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Fosfofrutoquinase-2/antagonistas & inibidores , Fosfofrutoquinase-2/metabolismo , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Piridinas/farmacologia , Pirróis/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Cell Metab ; 23(2): 280-91, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26774962

RESUMO

The oxygen-sensing prolyl hydroxylase domain proteins (PHDs) regulate cellular metabolism, but their role in neuronal metabolism during stroke is unknown. Here we report that PHD1 deficiency provides neuroprotection in a murine model of permanent brain ischemia. This was not due to an increased collateral vessel network. Instead, PHD1(-/-) neurons were protected against oxygen-nutrient deprivation by reprogramming glucose metabolism. Indeed, PHD1(-/-) neurons enhanced glucose flux through the oxidative pentose phosphate pathway by diverting glucose away from glycolysis. As a result, PHD1(-/-) neurons increased their redox buffering capacity to scavenge oxygen radicals in ischemia. Intracerebroventricular injection of PHD1-antisense oligonucleotides reduced the cerebral infarct size and neurological deficits following stroke. These data identify PHD1 as a regulator of neuronal metabolism and a potential therapeutic target in ischemic stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Reprogramação Celular , Deleção de Genes , Neurônios/metabolismo , Oxigênio/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Acidente Vascular Cerebral/prevenção & controle , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Isquemia Encefálica/complicações , Carbono/metabolismo , Reprogramação Celular/efeitos dos fármacos , Sequestradores de Radicais Livres/metabolismo , Hidroxilação , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Injeções Intraventriculares , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/farmacologia , Oxirredução/efeitos dos fármacos , Via de Pentose Fosfato/efeitos dos fármacos , Fenótipo , Pró-Colágeno-Prolina Dioxigenase/deficiência , Espécies Reativas de Oxigênio/metabolismo , Acidente Vascular Cerebral/complicações
13.
Nat Med ; 20(7): 741-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24929950

RESUMO

The N-terminal fragment of prolactin (16K PRL) inhibits tumor growth by impairing angiogenesis, but the underlying mechanisms are unknown. Here, we found that 16K PRL binds the fibrinolytic inhibitor plasminogen activator inhibitor-1 (PAI-1), which is known to contextually promote tumor angiogenesis and growth. Loss of PAI-1 abrogated the antitumoral and antiangiogenic effects of 16K PRL. PAI-1 bound the ternary complex PAI-1-urokinase-type plasminogen activator (uPA)-uPA receptor (uPAR), thereby exerting antiangiogenic effects. By inhibiting the antifibrinolytic activity of PAI-1, 16K PRL also protected mice against thromboembolism and promoted arterial clot lysis. Thus, by signaling through the PAI-1-uPA-uPAR complex, 16K PRL impairs tumor vascularization and growth and, by inhibiting the antifibrinolytic activity of PAI-1, promotes thrombolysis.


Assuntos
Fibrinólise , Neovascularização Patológica , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Prolactina/fisiologia , Animais , Divisão Celular , Células Cultivadas , Humanos , Camundongos , Camundongos Knockout , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/fisiologia , Prolactina/química
14.
Cell Metab ; 19(1): 37-48, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24332967

RESUMO

Strategies targeting pathological angiogenesis have focused primarily on blocking vascular endothelial growth factor (VEGF), but resistance and insufficient efficacy limit their success, mandating alternative antiangiogenic strategies. We recently provided genetic evidence that the glycolytic activator phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) promotes vessel formation but did not explore the antiangiogenic therapeutic potential of PFKFB3 blockade. Here, we show that blockade of PFKFB3 by the small molecule 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) reduced vessel sprouting in endothelial cell (EC) spheroids, zebrafish embryos, and the postnatal mouse retina by inhibiting EC proliferation and migration. 3PO also suppressed vascular hyperbranching induced by inhibition of Notch or VEGF receptor 1 (VEGFR1) and amplified the antiangiogenic effect of VEGF blockade. Although 3PO reduced glycolysis only partially and transiently in vivo, this sufficed to decrease pathological neovascularization in ocular and inflammatory models. These insights may offer therapeutic antiangiogenic opportunities.


Assuntos
Glicólise , Neovascularização Patológica/enzimologia , Fosfofrutoquinase-2/antagonistas & inibidores , Inibidores da Angiogênese/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/genética , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Fosfofrutoquinase-2/metabolismo , Piridinas/farmacologia , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/crescimento & desenvolvimento , Vasos Retinianos/patologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
15.
Proc Natl Acad Sci U S A ; 109(47): E3231-40, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23129614

RESUMO

Skeletal muscle regeneration mainly depends on satellite cells, a population of resident muscle stem cells. However, our understanding of the molecular mechanisms underlying satellite cell activation is still largely undefined. Here, we show that Cripto, a regulator of early embryogenesis, is a novel regulator of muscle regeneration and satellite cell progression toward the myogenic lineage. Conditional inactivation of cripto in adult satellite cells compromises skeletal muscle regeneration, whereas gain of function of Cripto accelerates regeneration, leading to muscle hypertrophy. Moreover, we provide evidence that Cripto modulates myogenic cell determination and promotes proliferation by antagonizing the TGF-ß ligand myostatin. Our data provide unique insights into the molecular and cellular basis of Cripto activity in skeletal muscle regeneration and raise previously undescribed implications for stem cell biology and regenerative medicine.


Assuntos
Linhagem da Célula , Fator de Crescimento Epidérmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Músculo Esquelético/fisiologia , Miostatina/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia , Envelhecimento/metabolismo , Animais , Proliferação de Células , Deleção de Genes , Marcação de Genes , Hipertrofia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Miostatina/metabolismo , Transdução de Sinais
16.
J Clin Invest ; 115(1): 118-27, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15630451

RESUMO

The angiogenic mechanism and therapeutic potential of PDGF-CC, a recently discovered member of the VEGF/PDGF superfamily, remain incompletely characterized. Here we report that PDGF-CC mobilized endothelial progenitor cells in ischemic conditions; induced differentiation of bone marrow cells into ECs; and stimulated migration of ECs. Furthermore, PDGF-CC induced the differentiation of bone marrow cells into smooth muscle cells and stimulated their growth during vessel sprouting. Moreover, delivery of PDGF-CC enhanced postischemic revascularization of the heart and limb. Modulating the activity of PDGF-CC may provide novel opportunities for treating ischemic diseases.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Isquemia/tratamento farmacológico , Isquemia/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Células-Tronco/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/citologia , Vasos Coronários/efeitos dos fármacos , Membro Posterior/irrigação sanguínea , Membro Posterior/efeitos dos fármacos , Humanos , Isquemia/induzido quimicamente , Isquemia/metabolismo , Linfocinas , Camundongos , Microcirculação/efeitos dos fármacos , Miocárdio/metabolismo , Miocárdio/patologia , Fosfotirosina/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Células-Tronco/citologia
17.
J Clin Invest ; 109(3): 327-36, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11827992

RESUMO

The murine VEGF gene is alternatively transcribed to yield the VEGF(120), VEGF(164), and VEGF(188) isoforms, which differ in their potential to bind to heparan sulfate and neuropilin-1 and to stimulate endothelial growth. Here, their role in retinal vascular development was studied in mice selectively expressing single isoforms. VEGF(164/164) mice were normal, healthy, and had normal retinal angiogenesis. In contrast, VEGF(120/120) mice exhibited severe defects in vascular outgrowth and patterning, whereas VEGF(188/188) mice displayed normal venular outgrowth but impaired arterial development. It is noteworthy that neuropilin-1, a receptor for VEGF(164), was predominantly expressed in retinal arterioles. These findings reveal distinct roles of the various VEGF isoforms in vascular patterning and arterial development in the retina.


Assuntos
Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/fisiologia , Linfocinas/genética , Linfocinas/fisiologia , Vasos Retinianos/crescimento & desenvolvimento , Animais , Arteríolas/anormalidades , Arteríolas/crescimento & desenvolvimento , Sequência de Bases , Padronização Corporal/genética , Padronização Corporal/fisiologia , DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Masculino , Camundongos , Camundongos Transgênicos , Gravidez , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Vasos Retinianos/anormalidades , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Vênulas/anormalidades , Vênulas/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA