Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Angiogenesis ; 25(3): 355-371, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35112158

RESUMO

Glioblastoma stem cells (GSCs) reside close to blood vessels (BVs) but vascular cues contributing to GSC stemness and the nature of GSC-BVs cross talk are not fully understood. Here, we dissected vascular cues influencing GSC gene expression and function to perfusion-based vascular cues, as well as to those requiring direct GSC-endothelial cell (EC) contacts. In light of our previous finding that perivascular tumor cells are metabolically different from tumor cells residing further downstream, cancer cells residing within a narrow, < 60 µm wide perivascular niche were isolated and confirmed to possess a superior tumor-initiation potential compared with those residing further downstream. To circumvent reliance on marker expression, perivascular GSCs were isolated from the respective locales based on their relative state of quiescence. Combined use of these procedures uncovered a large number of previously unrecognized differentially expressed GSC genes. We show that the unique metabolic milieu of the perivascular niche dominated by the highly restricted zone of mTOR activity is conducive for acquisition of GSC properties, primarily in the regulation of genes implicated in cell cycle control. A complementary role of vascular cues including those requiring direct glioma/EC contacts was revealed using glioma/EC co-cultures. Outstanding in the group of glioma cells impacted by nearby ECs were multiple genes responsible for maintaining GSCs in an undifferentiated state, a large fraction of which also relied on Notch-mediated signaling. Glioma-EC communication was found to be bidirectional, evidenced by extensive Notch-mediated EC reprogramming by contacting tumor cells, primarily metabolic EC reprogramming.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sinais (Psicologia) , Glioblastoma/patologia , Glioma/irrigação sanguínea , Glioma/genética , Humanos , Células-Tronco Neoplásicas/patologia
2.
Transl Stroke Res ; 13(5): 774-791, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35175562

RESUMO

The breakdown of the blood-brain barrier (BBB) is a critical event in the development of secondary brain injury after stroke. Among the cellular hallmarks in the acute phase after stroke are a downregulation of tight-junction molecules and the loss of microvascular pericyte coverage and endothelial sealing. Thus, a rapid repair of blood vessel integrity and re-stabilization of the BBB is considered an important strategy to reduce secondary brain damage. However, the mechanisms underlying BBB disruption remain poorly understood. Especially, the role of VEGF in this context remains inconclusive. With the conditional and reversible VEGF expression systems, we studied the time windows of deleterious and beneficial VEGF actions on blood vessel integrity in mice. Using genetic systems for gain of function and loss of function experiments, we activated and inhibited VEGF signaling prior and simultaneously to ischemic stroke onset. In both scenarios, VEGF seems to play a vital role in containing the stroke-induced damage after cerebral ischemia. We report that the transgenic overexpression of VEGF (GOF) prior to the stroke stabilizes the vasculature and prevents blood-brain barrier disruption in young and aged animals after stroke. Whereas inhibition of signals for endogenous VEGF (LOF) prior to stroke results in bigger infarction with massive brain swelling and enhanced BBB permeability, furthermore, activating or blocking VEGF signaling after ischemic stroke onset had comparable effects on BBB repair and cerebral edema. VEGF can function as an anti-permeability factor, and a VEGF-based therapy in the context of stroke prevention and recovery has an enormous potential.


Assuntos
Edema Encefálico , Isquemia Encefálica , AVC Isquêmico , Fator A de Crescimento do Endotélio Vascular , Animais , Barreira Hematoencefálica/metabolismo , Edema Encefálico/metabolismo , Isquemia Encefálica/complicações , AVC Isquêmico/complicações , Camundongos , Camundongos Transgênicos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Elife ; 92020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32496193

RESUMO

Blood vessels (BVs) are considered an integral component of neural stem cells (NSCs) niches. NSCs in the dentate gyrus (DG(have enigmatic elaborated apical cellular processes that are associated with BVs. Whether this contact serves as a mechanism for delivering circulating molecules is not known. Here we uncovered a previously unrecognized communication route allowing exclusive direct access of blood-borne substances to hippocampal NSCs. BBB-impermeable fluorescent tracer injected transcardially to mice is selectively uptaken by DG NSCs within a minute, via the vessel-associated apical processes. These processes, measured >30 nm in diameter, establish direct membrane-to-membrane contact with endothelial cells in specialized areas of irregular endothelial basement membrane and enriched with vesicular activity. Doxorubicin, a brain-impermeable chemotherapeutic agent, is also readily and selectively uptaken by NSCs and reduces their proliferation, which might explain its problematic anti-neurogenic or cognitive side-effect. The newly-discovered NSC-BV communication route explains how circulatory neurogenic mediators are 'sensed' by NSCs.


Assuntos
Células Endoteliais/citologia , Hipocampo/citologia , Células-Tronco Neurais/fisiologia , Animais , Antibióticos Antineoplásicos/metabolismo , Membrana Basal/citologia , Membrana Basal/metabolismo , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Comunicação Celular , Extensões da Superfície Celular/metabolismo , Extensões da Superfície Celular/fisiologia , Vesículas Citoplasmáticas/metabolismo , Doxorrubicina/metabolismo , Células Endoteliais/metabolismo , Feminino , Substâncias de Crescimento/metabolismo , Masculino , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese
4.
J Neurosci ; 40(5): 974-995, 2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-31959697

RESUMO

Multiple insults to the brain lead to neuronal cell death, thus raising the question to what extent can lost neurons be replenished by adult neurogenesis. Here we focused on the hippocampus and especially the dentate gyrus (DG), a vulnerable brain region and one of the two sites where adult neuronal stem cells (NSCs) reside. While adult hippocampal neurogenesis was extensively studied with regard to its contribution to cognitive enhancement, we focused on their underestimated capability to repair a massively injured, nonfunctional DG. To address this issue, we inflicted substantial DG-specific damage in mice of either sex either by diphtheria toxin-based ablation of >50% of mature DG granule cells (GCs) or by prolonged brain-specific VEGF overexpression culminating in extensive, highly selective loss of DG GCs (thereby also reinforcing the notion of selective DG vulnerability). The neurogenic system promoted effective regeneration by increasing NSCs proliferation/survival rates, restoring a nearly original DG mass, promoting proper rewiring of regenerated neurons to their afferent and efferent partners, and regaining of lost spatial memory. Notably, concomitantly with the natural age-related decline in the levels of neurogenesis, the regenerative capacity of the hippocampus also subsided with age. The study thus revealed an unappreciated regenerative potential of the young DG and suggests hippocampal NSCs as a critical reservoir enabling recovery from catastrophic DG damage.SIGNIFICANCE STATEMENT Adult hippocampal neurogenesis has been extensively studied in the context of its role in cognitive enhancement, but whether, and to what extent can dentate gyrus (DG)-resident neural stem cells drive regeneration of an injured DG has remained unclear. Here we show that DG neurogenesis acts to replace lost neurons and restore lost functions even following massive (>50%) neuronal loss. Age-related decline of neurogenesis is paralleled by a progressive decline of regenerative capacity. Considering also the exceptional vulnerability of the DG to insults, these findings provide a further rationale for maintaining DG neurogenesis in adult life.


Assuntos
Giro Denteado/fisiopatologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Proliferação de Células , Sobrevivência Celular , Giro Denteado/lesões , Giro Denteado/patologia , Feminino , Masculino , Camundongos Transgênicos
5.
Bio Protoc ; 10(10): e3628, 2020 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-33659301

RESUMO

Differential exposure of tumor cells to microenvironmental cues greatly impacts cell phenotypes, raising a need for position based sorting of tumor cells amenable to multiple OMICs and functional analyses. One such key determinant of tumor heterogeneity in solid tumors is its vasculature. Proximity to blood vessels (BVs) profoundly affects tumor cell phenotypes due to differential availability of oxygen, gradient exposure to blood-borne substances and inputs by angiocrine factors. To unravel the whole spectrum of genes, pathways and phenotypes impacted by BVs and to determine spatial domains of vascular influences, we developed a methodology for sorting tumor cells according to their relative distance from BVs. The procedure exemplified here using glioblastoma (GBM) model is based on differential uptake of intra-venously injected, freely-diffusing fluorescent dye that allows separation of stroma-free tumor cells residing in different, successive microenvironments amenable for subsequent OMICs and functional analyses. This reliable, easy to use, cost effective strategy can be extended to all solid tumors to study the impact of vasculature or the lack of it.

6.
Cell Metab ; 30(1): 201-211.e6, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31056286

RESUMO

Differential exposure of tumor cells to blood-borne and angiocrine factors results in diverse metabolic microenvironments conducive for non-genetic tumor cell diversification. Here, we harnessed a methodology for retrospective sorting of fully functional, stroma-free cancer cells solely on the basis of their relative distance from blood vessels (BVs) to unveil the whole spectrum of genes, metabolites, and biological traits impacted by BV proximity. In both grafted mouse tumors and natural human glioblastoma (GBM), mTOR activity was confined to few cell layers from the nearest perfused vessel. Cancer cells within this perivascular tier are distinguished by intense anabolic metabolism and defy the Warburg principle through exercising extensive oxidative phosphorylation. Functional traits acquired by perivascular cancer cells, namely, enhanced tumorigenicity, superior migratory or invasive capabilities, and, unexpectedly, exceptional chemo- and radioresistance, are all mTOR dependent. Taken together, the study revealed a previously unappreciated graded metabolic zonation directly impacting the acquisition of multiple aggressive tumor traits.


Assuntos
Glioblastoma/metabolismo , Metabolômica/métodos , Animais , Apoptose/fisiologia , Vasos Sanguíneos/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Tamanho Celular , Sobrevivência Celular/fisiologia , Citometria de Fluxo , Humanos , Immunoblotting , Masculino , Camundongos , Camundongos SCID , Mitocôndrias/metabolismo , Consumo de Oxigênio/fisiologia , Análise de Componente Principal
7.
Glia ; 67(4): 594-618, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30453385

RESUMO

Neurogenic roles of microglia (MG) are thought to include an active role in adult hippocampal neurogenesis in addition to their established roles in pruning surplus dendrites and clearing dead neuroblasts. However, identification of such a role and its delineation in the neurogenic cascade is yet to be established. Using diphtheria toxin-aided MG ablation, we show that MG reduction in the DG-the site where neuronal stem cells (NSCs) reside-is sufficient to impede overall hippocampal neurogenesis due to reduced survival of newly formed neuroblasts. To examine whether MG residing in the hippocampal neurogenic zone are inherently different from MG residing elsewhere in the hippocampus, we compared growth factor responsiveness of DG MG with that of CA1 MG. Strikingly, transgenic induction of the potent neurogenic factor VEGF elicited robust on-site MG expansion and activation exclusively in the DG and despite eliciting a comparable angiogenic response in the CA1 and elsewhere. Temporally, DG-specific MG expansion preceded both angiogenic and neurogenic responses. Remarkably, even partial MG reduction during the process of VEGF-induced neurogenesis led to reducing the number of newly formed neuroblasts to the basal level. Transcriptomic analysis of MG retrieved from the naïve DG and CA1 uncovered a set of genes preferentially expressed in DG MG. Notably the tyrosine kinase Axl is exclusively expressed in naïve and VEGF-induced DG MG and its inhibition prevented neurogenesis augmentation by VEGF. Taken together, findings uncover inherent unique properties of DG MG of supporting both basal- and VEGF-induced adult hippocampal neurogenesis.


Assuntos
Giro Denteado/citologia , Microglia/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Benzocicloeptenos/farmacologia , Vasos Sanguíneos/citologia , Transplante de Medula Óssea , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Caspase 3/metabolismo , Proliferação de Células , Desoxiuridina/farmacologia , Toxina Diftérica/toxicidade , Proteínas do Domínio Duplacortina , Inibidores Enzimáticos/farmacologia , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais/transplante , Neuropeptídeos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/metabolismo , Triazóis/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética
8.
J Exp Med ; 216(1): 215-230, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30545903

RESUMO

Insufficient erythropoiesis due to increased demand is usually met by hypoxia-driven up-regulation of erythropoietin (Epo). Here, we uncovered vascular endothelial growth factor (VEGF) as a novel inducer of Epo capable of increasing circulating Epo under normoxic, nonanemic conditions in a previously unrecognized reservoir of Epo-producing cells (EPCs), leading to expansion of the erythroid progenitor pool and robust splenic erythropoiesis. Epo induction by VEGF occurs in kidney, liver, and spleen in a population of Gli1+SMA+PDGFRß+ cells, a signature shared with vascular smooth muscle cells (VSMCs) derived from mesenchymal stem cell-like progenitors. Surprisingly, inhibition of PDGFRß signaling, but not VEGF signaling, abrogated VEGF-induced Epo synthesis. We thus introduce VEGF as a new player in Epo induction and perivascular Gli1+SMA+PDGFRß+ cells as a previously unrecognized EPC reservoir that could be harnessed for augmenting Epo synthesis in circumstances such as chronic kidney disease where production by canonical EPCs is compromised.


Assuntos
Eritropoese , Eritropoetina/biossíntese , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Hipóxia Celular , Células Precursoras Eritroides , Eritropoetina/genética , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/genética , Células Estromais/citologia , Células Estromais/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
9.
Nucleic Acids Res ; 46(21): 11396-11404, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30329087

RESUMO

The gene encoding the kinase Mnk2 (MKNK2) is alternatively spliced to produce two isoforms-Mnk2a and Mnk2b. We previously showed that Mnk2a is downregulated in several types of cancer and acts as a tumor suppressor by activation of the p38-MAPK stress pathway, inducing apoptosis. Moreover, Mnk2a overexpression suppressed Ras-induced transformation in culture and in vivo. In contrast, the Mnk2b isoform acts as a pro-oncogenic factor. In this study, we designed modified-RNA antisense oligonucleotides and screened for those that specifically induce a strong switch in alternative splicing of the MKNK2 gene (splice switching oligonucleotides or SSOs), elevating the tumor suppressive isoform Mnk2a at the expense of the pro-oncogenic isoform Mnk2b. Induction of Mnk2a by SSOs in glioblastoma cells activated the p38-MAPK pathway, inhibited the oncogenic properties of the cells, re-sensitized the cells to chemotherapy and inhibited glioblastoma development in vivo. Moreover, inhibition of p38-MAPK partially rescued glioblastoma cells suggesting that most of the anti-oncogenic activity of the SSO is mediated by activation of this pathway. These results suggest that manipulation of MKNK2 alternative splicing by SSOs is a novel approach to inhibit glioblastoma tumorigenesis.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Oligonucleotídeos/genética , Proteínas Serina-Treonina Quinases/genética , Processamento Alternativo , Animais , Apoptose , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Genes Supressores de Tumor , Glioblastoma/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Oligonucleotídeos Antissenso , Fosforilação , Isoformas de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Splicing de RNA , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Cell Metab ; 28(4): 573-587.e13, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30017355

RESUMO

The role of phosphoglycerate dehydrogenase (PHGDH), a key enzyme of the serine synthesis pathway (SSP), in endothelial cells (ECs) remains poorly characterized. We report that mouse neonates with EC-specific PHGDH deficiency suffer lethal vascular defects within days of gene inactivation, due to reduced EC proliferation and survival. In addition to nucleotide synthesis impairment, PHGDH knockdown (PHGDHKD) caused oxidative stress, due not only to decreased glutathione and NADPH synthesis but also to mitochondrial dysfunction. Electron transport chain (ETC) enzyme activities were compromised upon PHGDHKD because of insufficient heme production due to cellular serine depletion, not observed in other cell types. As a result of heme depletion, elevated reactive oxygen species levels caused EC demise. Supplementation of hemin in PHGDHKD ECs restored ETC function and rescued the apoptosis and angiogenesis defects. These data argue that ECs die upon PHGDH inhibition, even without external serine deprivation, illustrating an unusual importance of serine synthesis for ECs.


Assuntos
Células Endoteliais/metabolismo , Heme/metabolismo , Fosfoglicerato Desidrogenase/genética , Fosfoglicerato Desidrogenase/metabolismo , Serina/metabolismo , Apoptose , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Suplementos Nutricionais , Técnicas de Silenciamento de Genes , Hemina/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Microcefalia/metabolismo , Mitocôndrias/metabolismo , Mitofagia , Neovascularização Fisiológica , Estresse Oxidativo , Fosfoglicerato Desidrogenase/deficiência , Biossíntese de Proteínas , Transtornos Psicomotores/metabolismo , Purinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Convulsões/metabolismo
11.
Immunity ; 48(5): 951-962.e5, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29768178

RESUMO

Natural killer cells (NKs) are abundant in the human decidua, regulating trophoblast invasion and angiogenesis. Several diseases of poor placental development are associated with first pregnancies, so we thus looked to characterize differences in decidual NKs (dNKs) in first versus repeated pregnancies. We discovered a population found in repeated pregnancies, which has a unique transcriptome and epigenetic signature, and is characterized by high expression of the receptors NKG2C and LILRB1. We named these cells Pregnancy Trained decidual NK cells (PTdNKs). PTdNKs have open chromatin around the enhancers of IFNG and VEGFA. Activation of PTdNKs led to increased production and secretion of IFN-γ and VEGFα, with the latter supporting vascular sprouting and tumor growth. The precursors of PTdNKs seem to be found in the endometrium. Because repeated pregnancies are associated with improved placentation, we propose that PTdNKs, which are present primarily in repeated pregnancies, might be involved in proper placentation.


Assuntos
Memória Imunológica/imunologia , Células Matadoras Naturais/imunologia , Transcriptoma/imunologia , Útero/imunologia , Animais , Linhagem Celular Tumoral , Decídua/imunologia , Decídua/metabolismo , Feminino , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Células Matadoras Naturais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Gravidez , Útero/citologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Cell Rep ; 21(9): 2384-2392, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-29186678

RESUMO

Preexisting diabetes is a risk factor for the development of multiple types of cancer. Additionally, diabetic patients face a poorer prognosis when diagnosed with cancer. To gain insight into the effects of hyperglycemia, a hallmark of diabetes, on tumor growth and metastatic progression, we combined mouse models of cancer and hyperglycemia. We show that while hyperglycemia attenuates primary tumor growth, it concomitantly increases metastatic seeding in a distant organ. We further show that the increase in metastatic seeding is due to impaired secretion of granulocyte colony-stimulating factor (G-CSF) and impaired neutrophil mobilization. Normalizing blood glucose levels using insulin rescues neutrophil recruitment and tumor growth and concomitantly reduces metastatic seeding. These results provide links among hyperglycemia-induced changes in neutrophil mobilization, primary tumor growth, and metastatic progression. Furthermore, our observations highlight the importance of normalizing blood glucose levels in hyperglycemic cancer patients.


Assuntos
Hiperglicemia/metabolismo , Neutrófilos/fisiologia , Animais , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Feminino , Fator Estimulador de Colônias de Granulócitos/imunologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Hiperglicemia/imunologia , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica/imunologia
13.
Diabetologia ; 60(6): 1051-1056, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28299380

RESUMO

AIMS/HYPOTHESIS: Endothelial-endocrine cell interactions and vascular endothelial growth factor (VEGF)-A signalling are deemed essential for maternal islet vascularisation, glucose control and beta cell expansion during mouse pregnancy. The aim of this study was to assess whether pregnancy-associated beta cell expansion was affected under conditions of islet hypovascularisation. METHODS: Soluble fms-like tyrosine kinase 1 (sFLT1), a VEGF-A decoy receptor, was conditionally overexpressed in maternal mouse beta cells from 1.5 to 14.5 days post coitum. Islet vascularisation, glycaemic control, beta cell proliferation, individual beta cell size and total beta cell volume were assessed in both pregnant mice and non-pregnant littermates. RESULTS: Conditional overexpression of sFLT1 in beta cells resulted in islet hypovascularisation and glucose intolerance in both pregnant and non-pregnant mice. In contrast to non-pregnant littermates, glucose intolerance in pregnant mice was transient. sFLT1 overexpression did not affect pregnancy-associated changes in beta cell proliferation, individual beta cell size or total beta cell volume. CONCLUSIONS/INTERPRETATION: Reduced intra-islet VEGF-A signalling results in maternal islet hypovascularisation and impaired glycaemic control but does not preclude beta cell expansion during mouse pregnancy.


Assuntos
Células Secretoras de Insulina/metabolismo , Animais , Proliferação de Células/genética , Proliferação de Células/fisiologia , Tamanho Celular , Feminino , Ilhotas Pancreáticas/metabolismo , Camundongos , Gravidez , Ratos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
Proc Natl Acad Sci U S A ; 113(48): E7828-E7836, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27849577

RESUMO

Several factors are known to enhance adult hippocampal neurogenesis but a factor capable of inducing a long-lasting neurogenic enhancement that attenuates age-related neurogenic decay has not been described. Here, we studied hippocampal neurogenesis following conditional VEGF induction in the adult brain and showed that a short episode of VEGF exposure withdrawn shortly after the generation of durable new vessels (but not under conditions where newly made vessels failed to persist) is sufficient for neurogenesis to proceed at a markedly elevated level for many months later. Continual neurogenic increase over several months was not accompanied by accelerated exhaustion of the neuronal stem cell (NSC) reserve, thereby allowing neurogenesis to proceed at a markedly elevated rate also in old mice. Neurogenic enhancement by VEGF preconditioning was, in part, attributed to rescue of age-related NSC quiescence. Remarkably, VEGF caused extensive NSC remodelling manifested in transition of the enigmatic NSC terminal arbor onto long cytoplasmic processes engaging with and spreading over even remote blood vessels, a configuration reminiscent of early postnatal "juvenile" NSCs. Together, these findings suggest that VEGF preconditioning might be harnessed for long-term neurogenic enhancement despite continued exposure to an "aged" systemic milieu.


Assuntos
Envelhecimento , Giro Denteado/citologia , Células-Tronco Neurais/fisiologia , Neurogênese , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Forma Celular , Circulação Cerebrovascular , Giro Denteado/irrigação sanguínea , Giro Denteado/fisiologia , Feminino , Expressão Gênica , Masculino , Camundongos Transgênicos , Nicho de Células-Tronco
15.
Mech Dev ; 142: 40-49, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27794491

RESUMO

A fundamental issue in organogenesis is how dichotomous fate decisions are made securing proper allocation of multipotent progenitors to their respective descendants. Previous lineage tracing analyses showing Isl1+/VEGFR2+ cardiac progenitors in the second heart field give rise to both endocardium and myocardium suggest VEGF plays a role in this fate decision, conceivably promoting an endocardial fate. Isl1+ multipotent progenitors and lineage-committed descendants thereof were visualized and quantified within their transition zone in the outflow tract. Forced VEGF expression during the critical E8.5-E10.5 interval tilted the balance between myocardial- and endocardial-committed progenitors towards the latter, culminating in generation of surplus endocardium developing at the expense of a much thinner myocardium. Experiments ruled-out that surplus endocardium is due to VEGF-induced endocardial proliferation and that reduced myocardium is due to myocardial apoptosis. Inducing VEGF after most Isl1+ progenitors have been exhausted had no effect on the normal endocardia/myocardial ratio but instead produced an unrelated coronary phenotype. Together, these results uncover a novel role for VEGF in controlling proper allocation of Isl1+ cardiac progenitors to their respective descending lineages.


Assuntos
Proliferação de Células/genética , Proteínas com Homeodomínio LIM/genética , Organogênese/genética , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Apoptose/genética , Linhagem da Célula/genética , Endocárdio/citologia , Endocárdio/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/metabolismo , Células-Tronco/metabolismo
16.
Mech Dev ; 138 Pt 1: 56-62, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26103548

RESUMO

Blood vessels (BVs) not only serve as conduits for oxygen and nutrients but may also fulfill perfusion-independent functions. A growing body of data suggests that blood vessels are an integral component of stem cell niches, including stem cell niches in the adult brain. This review summarizes in vivo studies supporting the contention that blood vessels may indeed control function of neuronal stem cells (NSCs) residing in the two major neurogenic niches of the adult brain, namely the sub-ventricular zone and the hippocampus. The review discusses different modes of BV-NSC communication and possible mechanisms by which BV may modulate NSC behavior and responses to external stimuli.


Assuntos
Encéfalo/irrigação sanguínea , Neurogênese/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Humanos , Neurônios/fisiologia
18.
J Clin Invest ; 125(3): 1319-28, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25689256

RESUMO

Premature birth is a major risk factor for multiple brain pathologies, notably periventricular leukomalacia (PVL), which is distinguished by bilateral necrosis of neural tissue around the ventricles and a sequela of neurological disturbances. The 2 hallmarks of brain pathologies of prematurity are a restricted gestational window of vulnerability and confinement of injury to a specific cerebral region. Here, we examined the proposition that both of these features are determined by the state of blood vessel immaturity. We developed a murine genetic model that allows for inducible and reversible VEGF blockade during brain development. Using this system, we determined that cerebral vessels mature in a centrifugal, wave-like fashion that results in sequential acquisition of a functional blood-brain barrier and exit from a VEGF-dependent phase, with periventricular vessels being the last to mature. This developmental program permitted selective ablation of periventricular vessels via episodic VEGF blockade within a specific, vulnerable gestational window. Enforced collapse of ganglionic eminence vessels and resultant periventricular neural apoptosis resulted in a PVL-like phenotype that recapitulates the primary periventricular lesion, ventricular enlargement, and the secondary cortical deficit in out-migrating GABAergic inhibitory interneurons. These findings provide an animal model that reproduces the temporal and spatial specificities of PVL and indicate that damage to VEGF-dependent, immature periventricular vessels contributes to PVL development.


Assuntos
Leucomalácia Periventricular/fisiopatologia , Neovascularização Fisiológica , Animais , Apoptose , Barreira Hematoencefálica , Hipóxia Celular , Ventrículos Cerebrais/irrigação sanguínea , Ventrículos Cerebrais/patologia , Modelos Animais de Doenças , Feminino , Humanos , Interneurônios/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Neurônios/fisiologia , Gravidez , Nascimento Prematuro/fisiopatologia , Transcrição Gênica , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
19.
PLoS One ; 9(3): e92869, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24667808

RESUMO

RATIONALE: Rescuing adverse myocardial remodeling is an unmet clinical goal and, correspondingly, pharmacological means for its intended reversal are urgently needed. OBJECTIVES: To harness a newly-developed experimental model recapitulating progressive heart failure development for the discovery of new drugs capable of reversing adverse remodeling. METHODS AND RESULTS: A VEGF-based conditional transgenic system was employed in which an induced perfusion deficit and a resultant compromised cardiac function lead to progressive remodeling and eventually heart failure. Ability of candidate drugs administered at sequential remodeling stages to reverse hypertrophy, enlarged LV size and improve cardiac function was monitored. Arguing for clinical relevance of the experimental system, clinically-used drugs operating on the Renin-Angiotensin-Aldosterone-System (RAAS), namely, the ACE inhibitor Enalapril and the direct renin inhibitor Aliskerin fully reversed remodeling. Remodeling reversal by these drugs was not accompanied by neovascularization and reached a point-of-no-return. Similarly, the PPARγ agonist Pioglitazone was proven capable of reversing all aspects of cardiac remodeling without affecting the vasculature. Extending the arsenal of remodeling-reversing drugs to pathways other than RAAS, a specific inhibitor of 11ß-hydroxy-steroid dehydrogenase type 1 (11ß HSD1), a key enzyme required for generating active glucocorticoids, fully rescued myocardial hypertrophy. This was associated with mitigating the hypertrophy-associated gene signature, including reversing the myosin heavy chain isoform switch but in a pattern distinguishable from that associated with neovascularization-induced reversal. CONCLUSIONS: A system was developed suitable for identifying novel remodeling-reversing drugs operating in different pathways and for gaining insights into their mechanisms of action, exemplified here by uncoupling their vascular affects.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , Cardiomegalia/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Sistema Renina-Angiotensina/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Animais , Cardiomegalia/enzimologia , Cardiomegalia/genética , Avaliação Pré-Clínica de Medicamentos , Camundongos , Camundongos Transgênicos , Sistema Renina-Angiotensina/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Diabetologia ; 57(1): 140-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24121626

RESUMO

AIMS/HYPOTHESIS: Vascular endothelial growth factor (VEGF) has been recognised by loss-of-function experiments as a pleiotropic factor with importance in embryonic pancreas development and postnatal beta cell function. Chronic, nonconditional overexpression of VEGF-A has a deleterious effect on beta cell development and function. We report, for the first time, a conditional gain-of-function study to evaluate the effect of transient VEGF-A overexpression by adult pancreatic beta cells on islet vasculature and beta cell proliferation and survival, under both normal physiological and injury conditions. METHODS: In a transgenicmouse strain, overexpressing VEGF-A in a doxycycline-inducible and beta cell-specific manner, we evaluated the ability of VEGF-A to affect islet vessel density, beta cell proliferation and protection of the adult beta cell mass from toxin-induced injury. RESULTS: Short-term VEGF-A overexpression resulted in islet hypervascularisation, increased beta cell proliferation and protection from toxin-mediated beta cell death, and thereby prevented the development of hyperglycaemia. Extended overexpression of VEGF-A led to impaired glucose tolerance, elevated fasting glycaemia and a decreased beta cell mass. CONCLUSIONS/INTERPRETATION: Overexpression of VEGF-A in beta cells time-dependently affects glycometabolic control and beta cell protection and proliferation. These data nourish further studies to examine the role of controlled VEGF delivery in (pre)clinical applications aimed at protecting and/or restoring the injured beta cell mass.


Assuntos
Diabetes Mellitus/prevenção & controle , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular/fisiologia , Diabetes Mellitus/metabolismo , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , Ratos , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA