Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Mol Metab ; 88: 102008, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39142562

RESUMO

OBJECTIVE: Maternal obesity is increasingly common and negatively impacts offspring health. Children of mothers with obesity are at higher risk of developing diseases linked to hematopoietic system abnormalities and metabolism such as type 2 diabetes. Interestingly, disease risks are often dependent on the offspring's sex, suggesting sex-specific reprogramming effect of maternal obesity on offspring hematopoietic stem and progenitor cell (HSPC) function. However, the impact of maternal obesity exposure on offspring HSPC function, and the capability of HSPC to regulate offspring metabolic health is largely understudied. This study aims to test the hypothesis that offspring of obese mice exhibit sex-differences in HSPC function that affect offspring's metabolic health. METHODS: We first assessed bone marrow hematopoietic stem and progenitor cell phenotype using postnatal day 21 (P21) and 8-week-old C57BL/6J mice born to control and diet-induced obese dams. We also sorted HSPC (Lineage-, Sca1+, cKit + cells) from P21 mice for competitive primary and secondary transplant, as well as transcriptomic analysis. Body weight, adiposity, insulin tolerance test and glucose tolerance tests were performed in primary and secondary transplant recipient animals. RESULTS: We discovered sex-differences in offspring HSPC function in response to maternal obesity exposure, where male offspring of obese dams (MatOb) showed decreased HSPC numbers and engraftment, while female MatOb offspring remained largely unaffected. RNA-seq revealed immune stimulatory pathways in female MatOb offspring. Finally, only recipients of male MatOb offspring HSPC exhibited glucose intolerance. CONCLUSIONS: This study demonstrated the lasting effect of maternal obesity exposure on offspring HSPC function and implicates HSPC in metabolic regulation.


Assuntos
Intolerância à Glucose , Células-Tronco Hematopoéticas , Camundongos Endogâmicos C57BL , Animais , Intolerância à Glucose/metabolismo , Feminino , Camundongos , Masculino , Células-Tronco Hematopoéticas/metabolismo , Gravidez , Obesidade Materna/metabolismo , Camundongos Obesos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Obesidade/metabolismo , Dieta Hiperlipídica/efeitos adversos
2.
J Clin Invest ; 134(12)2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38950330

RESUMO

Activating mutations of FLT3 contribute to deregulated hematopoietic stem and progenitor cell (HSC/Ps) growth and survival in patients with acute myeloid leukemia (AML), leading to poor overall survival. AML patients treated with investigational drugs targeting mutant FLT3, including Quizartinib and Crenolanib, develop resistance to these drugs. Development of resistance is largely due to acquisition of cooccurring mutations and activation of additional survival pathways, as well as emergence of additional FLT3 mutations. Despite the high prevalence of FLT3 mutations and their clinical significance in AML, there are few targeted therapeutic options available. We have identified 2 novel nicotinamide-based FLT3 inhibitors (HSN608 and HSN748) that target FLT3 mutations at subnanomolar concentrations and are potently effective against drug-resistant secondary mutations of FLT3. These compounds show antileukemic activity against FLT3ITD in drug-resistant AML, relapsed/refractory AML, and in AML bearing a combination of epigenetic mutations of TET2 along with FLT3ITD. We demonstrate that HSN748 outperformed the FDA-approved FLT3 inhibitor Gilteritinib in terms of inhibitory activity against FLT3ITD in vivo.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Leucemia Mieloide Aguda , Niacinamida , Tirosina Quinase 3 Semelhante a fms , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Animais , Camundongos , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Feminino , Antineoplásicos/farmacologia , Antineoplásicos/química , Mutação , Camundongos SCID , Camundongos Endogâmicos NOD
3.
Sci Rep ; 10(1): 17319, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33057033

RESUMO

Multiple Myeloma (MM) induces bone destruction, decreases bone formation, and increases marrow angiogenesis in patients. We reported that osteocytes (Ocys) directly interact with MM cells to increase tumor growth and expression of Ocy-derived factors that promote bone resorption and suppress bone formation. However, the contribution of Ocys to enhanced marrow vascularization in MM is unclear. Since the MM microenvironment is hypoxic, we assessed if hypoxia and/or interactions with MM cells increases pro-angiogenic signaling in Ocys. Hypoxia and/or co-culture with MM cells significantly increased Vegf-a expression in MLOA5-Ocys, and conditioned media (CM) from MLOA5s or MM-MLOA5 co-cultured in hypoxia, significantly increased endothelial tube length compared to normoxic CM. Further, Vegf-a knockdown in MLOA5s or primary Ocys co-cultured with MM cells or neutralizing Vegf-a in MM-Ocy co-culture CM completely blocked the increased endothelial activity. Importantly, Vegf-a-expressing Ocy numbers were significantly increased in MM-injected mouse bones, positively correlating with tumor vessel area. Finally, we demonstrate that direct contact with MM cells increases Ocy Fgf23, which enhanced Vegf-a expression in Ocys. Fgf23 deletion in Ocys blocked these changes. These results suggest hypoxia and MM cells induce a pro-angiogenic phenotype in Ocys via Fgf23 and Vegf-a signaling, which can promote MM-induced marrow vascularization.


Assuntos
Medula Óssea/irrigação sanguínea , Fatores de Crescimento de Fibroblastos/fisiologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Neovascularização Patológica/genética , Osteócitos/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Reabsorção Óssea/etiologia , Linhagem Celular , Feminino , Fator de Crescimento de Fibroblastos 23 , Expressão Gênica/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Osteócitos/metabolismo , Osteogênese , Microambiente Tumoral
4.
J Infect Dis ; 219(7): 1076-1083, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30239747

RESUMO

BACKGROUND: Human immunodeficiency virus (HIV) may be related to cardiovascular disease through monocyte activation-associated endothelial dysfunction. METHODS: Blood samples from 15 HIV-negative participants (the uninfected group), 8 HIV-positive participants who were not receiving antiretroviral therapy (ART) (the infected, untreated group), and 15 HIV-positive participants who were receiving ART (the infected, treated group) underwent flow cytometry of endothelial colony-forming cells (ECFCs) and monocyte proportions. IncuCyte live cell imaging of 8 capillary proliferative capacity parameters were obtained from cord blood ECFCs treated with participant plasma. RESULTS: The ECFC percentage determined by flow cytometry was not different between the study groups; however, values of the majority of capillary proliferative capacity parameters (ie, cell area, network length, network branch points, number of networks, and average tube width uniformity) were significantly lower in infected, untreated participants as compared to values for uninfected participants or infected, treated participants (P < .00625 for all comparisons). CD14+CD16+ intermediate monocytes and soluble CD163 were significantly and negatively correlated with several plasma-treated, cord blood ECFC proliferative capacity parameters in the combined HIV-positive groups but not in the uninfected group. CONCLUSIONS: Cord blood ECFC proliferative capacity was significantly impaired by plasma from infected, untreated patients, compared with plasma from uninfected participants and from infected, treated participants. Several ECFC functional parameters were adversely associated with monocyte activation in the HIV-positive groups, thereby suggesting a mechanism by which HIV-related inflammation may impair vascular reparative potential and consequently increase the risk of cardiovascular disease during HIV infection.


Assuntos
Endotélio/imunologia , Soronegatividade para HIV/imunologia , Soropositividade para HIV/imunologia , Monócitos , Células-Tronco , Adulto , Alcinos , Fármacos Anti-HIV/uso terapêutico , Benzoxazinas/uso terapêutico , Proliferação de Células , Quimiocina CCL5/sangue , Ciclopropanos , Endotélio/patologia , Feminino , Sangue Fetal , Citometria de Fluxo , Proteínas Ligadas por GPI/metabolismo , Soropositividade para HIV/sangue , Soropositividade para HIV/tratamento farmacológico , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Neovascularização Fisiológica , Plasma/imunologia , Estudos Prospectivos , Receptores de IgG/metabolismo , Células-Tronco/fisiologia , Molécula 1 de Adesão de Célula Vascular/sangue , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/sangue
5.
PLoS One ; 13(3): e0193749, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29538431

RESUMO

OBJECTIVE: Endothelial dysfunction is central to the pathogenesis of many rheumatic diseases, typified by vascular inflammation and damage. Immunosuppressive drugs induce disease remission and lead to improved patient survival. However, there remains a higher incidence of cardiovascular disease in these patients even after adequate disease control. The purpose of this study was to determine the effect of mycophenolic acid (MPA), a commonly used immunosuppressive drug in rheumatology, on blood vessel or circulating endothelial colony forming cell number and function. METHODS: We tested whether mycophenolic acid exerts an inhibitory effect on proliferation, clonogenic potential and vasculogenic function of endothelial colony forming cell. We also studied potential mechanisms involved in the observed effects. RESULTS: Treatment with MPA decreased endothelial colony forming cell proliferation, clonogenic potential and vasculogenic function in a dose-dependent fashion. MPA increased senescence-associated ß-galactosidase expression, p21 gene expression and p53 phosphorylation, indicative of activation of cellular senescence. Exogenous guanosine supplementation rescued diminished endothelial colony forming cell proliferation and indices of senescence, consistent with the known mechanism of action of MPA. CONCLUSION: Our findings show that clinically relevant doses of MPA have potent anti-angiogenic and pro-senescent effects on vascular precursor cells in vitro, thus indicating that treatment with MPA can potentially affect vascular repair and regeneration. This warrants further studies in vivo to determine how MPA therapy contributes to vascular dysfunction and increased cardiovascular disease seen in patients with inflammatory rheumatic disease.


Assuntos
Senescência Celular/efeitos dos fármacos , Ácido Micofenólico/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Galactosidases/metabolismo , Guanosina/farmacologia , Humanos , Fosforilação/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Cordão Umbilical/citologia
6.
Biol Blood Marrow Transplant ; 21(10): 1739-45, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26172478

RESUMO

Reliable, noninvasive methods for diagnosing and prognosing sinusoidal obstruction syndrome (SOS) early after hematopoietic cell transplantation (HCT) are needed. We used a quantitative mass spectrometry-based proteomics approach to identify candidate biomarkers of SOS by comparing plasma pooled from 20 patients with and 20 patients without SOS. Of 494 proteins quantified, we selected 6 proteins (L-Ficolin, vascular cell adhesion molecule-1 [VCAM1], tissue inhibitor of metalloproteinase-1, von Willebrand factor, intercellular adhesion molecule-1, and CD97) based on a differential heavy/light isotope ratio of at least 2 fold, information from the literature, and immunoassay availability. Next, we evaluated the diagnostic potential of these 6 proteins and 5 selected from the literature (suppression of tumorigenicity-2 [ST2], angiopoietin-2 (ANG2), hyaluronic acid [HA], thrombomodulin, and plasminogen activator inhibitor-1) in samples from 80 patients. The results demonstrate that together ST2, ANG2, L-Ficolin, HA, and VCAM1 compose a biomarker panel for diagnosis of SOS. L-Ficolin, HA, and VCAM1 also stratified patients at risk for SOS as early as the day of HCT. Prognostic Bayesian modeling for SOS onset based on L-Ficolin, HA, and VCAM1 levels on the day of HCT and clinical characteristics showed >80% correct prognosis of SOS onset. These biomarkers may provide opportunities for preemptive intervention to minimize SOS incidence and/or severity.


Assuntos
Biomarcadores/sangue , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Hepatopatia Veno-Oclusiva/diagnóstico , Ácido Hialurônico/sangue , Lectinas/sangue , Molécula 1 de Adesão de Célula Vascular/sangue , Adolescente , Adulto , Teorema de Bayes , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Hepatopatia Veno-Oclusiva/sangue , Hepatopatia Veno-Oclusiva/mortalidade , Humanos , Molécula 1 de Adesão Intercelular/sangue , Masculino , Espectrometria de Massas/métodos , Pessoa de Meia-Idade , Prognóstico , Proteômica , Medição de Risco , Inibidor Tecidual de Metaloproteinase-1/sangue , Adulto Jovem , Fator de von Willebrand/análise , Ficolinas
7.
Pediatr Pulmonol ; 50(12): 1313-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25462113

RESUMO

RATIONALE: While infants who are born extremely premature and develop bronchopulmonary dysplasia (BPD) have impaired alveolar development and decreased pulmonary diffusion (DLCO), it remains unclear whether infants born less premature and do not develop BPD, healthy premature (HP), have impaired parenchymal development. In addition, there is increasing evidence that pro-angiogenic cells are important for vascular development; however, there is little information on the relationship of pro-angiogenic cells to lung growth and development in infants. OBJECTIVE: and Methods Determine among healthy premature (HP) and fullterm (FT) infants, whether DLCO and alveolar volume (VA) are related to gestational age at birth (GA), respiratory support during the neonatal period (mechanical ventilation [MV], supplemental oxygen [O2], continuous positive airway pressure [CPAP]), and pro-angiogenic circulating hematopoietic stem/progenitor cells (CHSPCs). We measured DLCO, VA, and CHSPCs in infants between 3-33 months corrected-ages; HP (mean GA = 31.7 wks; N = 48,) and FT (mean GA = 39.3 wks; N =88). RESULT: DLCO was significantly higher in HP than FT subjects, while there was no difference in VA , after adjusting for body length, gender, and race. DLCO and VA were not associated with GA, MV and O2; however, higher values were associated with higher CHSPCs, as well as treatment with CPAP. CONCLUSION: Our findings suggest that in the absence of extreme premature birth, as well as BPD, prematurity per se, does not impair lung parenchymal development.


Assuntos
Recém-Nascido Prematuro/fisiologia , Pulmão/crescimento & desenvolvimento , Pressão Positiva Contínua nas Vias Aéreas , Feminino , Idade Gestacional , Células-Tronco Hematopoéticas/fisiologia , Humanos , Lactente , Recém-Nascido , Masculino , Células-Tronco/fisiologia , Nascimento a Termo
8.
Pediatr Res ; 77(2): 316-25, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25412163

RESUMO

BACKGROUND: Children exposed to gestational diabetes mellitus (GDM) during pregnancy are at increased risk of obesity, diabetes, and hypertension. Our goal was to identify metabolic and hematopoietic alterations after intrauterine exposure to maternal hyperglycemia that may contribute to the pathogenesis of chronic morbidities. METHODS: Streptozotocin treatment induced maternal hyperglycemia during the last third of gestation in rat dams. Offspring of control mothers (OCM) and diabetic mothers (ODM) were evaluated for weight, glucose tolerance, insulin tolerance, and hematopoiesis defects. The effects of aging were examined in normal and high-fat diet (HFD)-fed young (8-wk-old) and aged (11-mo-old) OCM and ODM rats. RESULTS: Young adult ODM males on a normal diet, but not females, displayed improved glucose tolerance due to increased insulin levels. Aged ODM males and females gained more weight than OCM on a HFD and had worse glucose tolerance. Aged ODM males fed a HFD were also neutrophilic. Increases in bone marrow cellularity and myeloid progenitors preceded neutrophilia in ODM males fed a HFD. CONCLUSION: When combined with other risk factors like HFD and aging, changes in glucose metabolism and hematopoiesis may contribute to the increased risk of obesity, type 2 diabetes, and hypertension observed in children of GDM mothers.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Gestacional/metabolismo , Doenças Fetais/metabolismo , Hematopoese/fisiologia , Fatores Etários , Animais , Peso Corporal/fisiologia , Feminino , Doenças Fetais/etiologia , Teste de Tolerância a Glucose , Resistência à Insulina/fisiologia , Masculino , Gravidez , Ratos , Fatores de Risco
9.
J Biol Chem ; 287(13): 10435-10443, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22315228

RESUMO

Loss-of-function mutation of Jup has been associated with Naxos disease, which is characterized by arrhythmogenic cardiomyopathy and the cutaneous disorder palmoplantar keratoderma. Previously, we have shown that genetic ablation of Jup in cardiomyocytes in mice leads to arrhythmogenic cardiomyopathy similar to Naxos disease in humans. Currently, to determine the pathogenesis of Naxos disease-associated keratoderma, we generated Jup mutant mice by inactivating Jup restrictively in keratinocytes. Jup mutant mice largely recapitulated the clinical features of human palmoplantar keratoderma: overcornification and thickening of the epidermis. Jup mutant mice also suffered skin ulceration and inflammation. Cell apoptosis and proliferation were significantly elevated in Jup mutant epidermis. Ultrastructural analyses revealed the disruption of the assembly of desmosomes and adherens junctions in Jup mutant epidermis. We also demonstrated the compensational increase in ß-catenin at Jup mutant cell-cell junctions without altering its signaling activities. Our findings provide important insights for understanding the pathogenesis of human palmoplantar keratoderma.


Assuntos
Apoptose , Displasia Arritmogênica Ventricular Direita/metabolismo , Proliferação de Células , Desmoplaquinas/metabolismo , Epiderme/metabolismo , Doenças do Cabelo/metabolismo , Ceratodermia Palmar e Plantar/metabolismo , gama Catenina/metabolismo , Junções Aderentes/genética , Junções Aderentes/metabolismo , Junções Aderentes/ultraestrutura , Animais , Displasia Arritmogênica Ventricular Direita/genética , Displasia Arritmogênica Ventricular Direita/patologia , Desmoplaquinas/genética , Desmossomos/genética , Desmossomos/metabolismo , Desmossomos/ultraestrutura , Epiderme/ultraestrutura , Doenças do Cabelo/genética , Doenças do Cabelo/patologia , Humanos , Ceratodermia Palmar e Plantar/genética , Ceratodermia Palmar e Plantar/patologia , Camundongos , Camundongos Transgênicos , gama Catenina/genética
10.
J Leukoc Biol ; 91(2): 333-40, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22106009

RESUMO

FA is a genetic disorder characterized by BM failure, developmental defects, and cancer predisposition. Previous studies suggest that FA patients exhibit alterations in immunologic function. However, it is unclear whether the defects are immune cell-autonomous or secondary to leukopenia from evolving BM failure. Given the central role that macrophages have in the innate immune response, inflammation resolution, and antigen presentation for acquired immunity, we examined whether macrophages from Fancc-/- mice exhibit impaired function. Peritoneal inflammation induced by LPS or sodium periodate resulted in reduced monocyte/macrophage recruitment in Fancc-/- mice compared with WT controls. Fancc-/- mice also had decreased inflammatory monocytes mobilized into the peripheral blood after LPS treatment compared with controls. Furthermore, Fancc-/- peritoneal macrophages displayed cell-autonomous defects in function, including impaired adhesion to FN or endothelial cells, reduced chemoattractant-mediated migration, and decreased phagocytosis. Moreover, dysregulated F-actin rearrangement was detected in Fancc-/- macrophages after adhesion to FN, which was consistent with an observed reduction in RhoA-GTP levels. Importantly, these data suggest that impaired cytoskeletal rearrangements in Fancc-/- macrophages may be the common mechanism responsible for cell-autonomous defects detected in vitro, as well as altered monocyte/macrophage trafficking in vivo.


Assuntos
Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Anemia de Fanconi/patologia , Macrófagos Peritoneais/fisiologia , Actinas/análise , Animais , Adesão Celular , Células Cultivadas/patologia , Quimiotaxia/efeitos dos fármacos , Técnicas de Cocultura , Citoesqueleto/química , Citoesqueleto/ultraestrutura , Células Endoteliais/citologia , Anemia de Fanconi/genética , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Proteína do Grupo de Complementação C da Anemia de Fanconi/fisiologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Modelos Animais , Fagocitose/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Superóxidos/metabolismo , Proteínas rho de Ligação ao GTP/fisiologia , Proteína rhoA de Ligação ao GTP
11.
Hum Mol Genet ; 20(23): 4582-96, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21880664

RESUMO

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inheritable myocardial disorder associated with fibrofatty replacement of myocardium and ventricular arrhythmia. A subset of ARVC is categorized as Naxos disease, which is characterized by ARVC and a cutaneous disorder. A homozygous loss-of-function mutation of the Plakoglobin (Jup) gene, which encodes a major component of the desmosome and the adherens junction, had been identified in Naxos patients, although the underlying mechanism remained elusive. We generated Jup mutant mice by ablating Jup in cardiomyocytes. Jup mutant mice largely recapitulated the clinical manifestation of human ARVC: ventricular dilation and aneurysm, cardiac fibrosis, cardiac dysfunction and spontaneous ventricular arrhythmias. Ultra-structural analyses revealed that desmosomes were absent in Jup mutant myocardia, whereas adherens junctions and gap junctions were preserved. We found that ventricular arrhythmias were associated with progressive cardiomyopathy and fibrosis in Jup mutant hearts. Massive cell death contributed to the cardiomyocyte dropout in Jup mutant hearts. Despite the increase of ß-catenin at adherens junctions in Jup mutant cardiomyoicytes, the Wnt/ß-catenin-mediated signaling was not altered. Transforming growth factor-beta-mediated signaling was found significantly elevated in Jup mutant cardiomyocytes at the early stage of cardiomyopathy, suggesting an important pathogenic pathway for Jup-related ARVC. These findings have provided further insights for the pathogenesis of ARVC and potential therapeutic interventions.


Assuntos
Displasia Arritmogênica Ventricular Direita/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , gama Catenina/deficiência , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/fisiopatologia , Displasia Arritmogênica Ventricular Direita/complicações , Displasia Arritmogênica Ventricular Direita/fisiopatologia , Morte Celular , Desmossomos/metabolismo , Desmossomos/ultraestrutura , Fibrose , Deleção de Genes , Sistema de Condução Cardíaco/patologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Camundongos , Camundongos Mutantes , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/ultraestrutura , Especificidade de Órgãos , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Via de Sinalização Wnt , gama Catenina/metabolismo
12.
J Cell Biochem ; 112(9): 2383-91, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21520247

RESUMO

FANCD2, a key factor in the FANC-BRCA1 pathway is monoubiquitinated and targeted to discrete nuclear foci following DNA damage. Since monoubiquitination of FANCD2 is a crucial indicator for cellular response to DNA damage, we monitored the fate of FANCD2 and its monoubiquitination following DNA damage. Disappearance of FANCD2 protein was induced following DNA damage in a dose-dependent manner, which correlated with degradation of BRCA1 and poly-ADP ribose polymerase (PARP), known targets for caspase-mediated apoptosis. Disappearance of FANCD2 was not affected by a proteasome inhibitor but was blocked by a caspase inhibitor. DNA damage-induced disappearance of FANCD2 was also observed in cells lacking FANCA, suggesting that disappearance of FANCD2 does not depend on FANC-BRCA1 pathway and FANCD2 monoubiquitination. In keeping with this, cells treated with TNF-α, an apoptotic stimulus without causing any DNA damage, also induced disappearance of FANCD2 without monoubiquitination. Together, our data suggest that FANCD2 is a target for caspase-mediated apoptotic pathway, which may be an early indicator for apoptotic cell death.


Assuntos
Apoptose , Caspases/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Cisplatino/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , Dano ao DNA , Células HeLa , Humanos , Mitomicina/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Ubiquitinadas/farmacologia , Proteínas Ubiquitinadas/fisiologia
13.
J Pediatr ; 157(4): 540-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20542287

RESUMO

OBJECTIVE: To evaluate whether counts of circulating colony forming unit-endothelial cells (CFU-ECs), cells co-expressing CD34, CD133, and CD31 (CD34+CD133+CD31+), and CD34+CD45- cells are altered in adolescents with type 1 diabetes and if the changes in counts correlate with endothelial dysfunction. STUDY DESIGN: Adolescents with diabetes (ages 18 to 22 years) and race- and sex-matched control subjects were studied. We assessed circulating CFU-ECs, using colony assays, and CD34+CD133+CD31+ and CD34+CD45- cells, using poly-chromatic flow cytometry. CFU-ECs and CD34+CD133+CD31+ are hematopoietic-derived progenitors that inversely correlate with cardiovascular risk in adults. CD34+CD45- cells are enriched for endothelial cells with robust vasculogenic potential. Vascular reactivity was tested by laser Doppler iontophoresis. RESULTS: Subjects with diabetes had lower CD34+CD133+CD31+ cells, a trend toward reduced CFU-ECs, and increased CD34+CD45- cells compared with control subjects. Endothelium-dependent vasodilation was impaired in subjects with diabetes, which correlated with reductions in circulating CD34+CD133+CD31+ cells. CONCLUSIONS: Long-term sequelae of type 1 diabetes include vasculopathies. Endothelial progenitor cells promote vascular health by facilitating endothelial integrity and function. Lower CD34+CD133+CD31+ cells may be a harbinger of future macrovascular disease risk. Higher circulating CD34+CD45- cells may reflect ongoing endothelial damage. These cells are potential biomarkers to guide therapeutic interventions to enhance endothelial function and to prevent progression to overt vascular disease.


Assuntos
Antígenos CD/imunologia , Biomarcadores/sangue , Diabetes Mellitus Tipo 1/fisiopatologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Adolescente , Velocidade do Fluxo Sanguíneo , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Endotélio Vascular/imunologia , Feminino , Humanos , Masculino , Vasodilatação/fisiologia , Adulto Jovem
14.
Blood ; 113(12): 2655-60, 2009 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-19168785

RESUMO

The underlying molecular mechanisms that promote bone marrow failure in Fanconi anemia are incompletely understood. Evidence suggests that enhanced apoptosis of hematopoietic precursors is a major contributing factor. Previously, enhanced apoptosis of Fanconi anemia type C-deficient (Fancc(-/-)) progenitors was shown to involve aberrant p38 MAPK activation. Given the importance of c-Jun N-terminal kinase (JNK) in the stress response, we tested whether enhanced apoptosis of Fancc(-/-) cells also involved altered JNK activation. In Fancc(-/-) murine embryonic fibroblasts, tumor necrosis factor alpha (TNF-alpha) induced elevated JNK activity. In addition, JNK inhibition protected Fancc(-/-) murine embryonic fibroblasts and c-kit(+) bone marrow cells from TNF-alpha-induced apoptosis. Importantly, hematopoietic progenitor assays demonstrated that JNK inhibition enhanced Fancc(-/-) colony formation in the presence of TNF-alpha. Competitive repopulation assays showed that Fancc(-/-) donor cells cultured with the JNK inhibitor had equivalent levels of donor chimerism compared with Fancc(-/-) donor cells cultured with vehicle control. In contrast, culturing Fancc(-/-) cells with a p38 MAPK inhibitor significantly increased repopulating ability, supporting an integral role of p38 MAPK in maintaining Fancc(-/-) hematopoietic stem cell function. Taken together, these data suggest that p38 MAPK, but not JNK, has a critical role in maintaining the engraftment of Fancc(-/-)-reconstituting cells under conditions of stress.


Assuntos
Proteína do Grupo de Complementação C da Anemia de Fanconi/deficiência , Anemia de Fanconi/enzimologia , Proteína Quinase 8 Ativada por Mitógeno/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Células Cultivadas/transplante , Ensaio de Unidades Formadoras de Colônias , Ativação Enzimática , Anemia de Fanconi/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/patologia , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/genética , Modelos Animais , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Quimera por Radiação , Estresse Fisiológico , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
15.
Am J Physiol Heart Circ Physiol ; 296(3): H877-86, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19151256

RESUMO

While tissue perfusion and angiogenesis subsequent to acute femoral artery occlusion are suppressed in NADPH oxidase 2 (Nox2)-null (Nox2(-/-)) mice, studies have not established the role of Nox2 in collateral artery enlargement. Rac2 is a small GTPase that binds Nox2 and activates Nox2-based NAD(P)H oxidase but, unlike Nox2, is primarily restricted to bone marrow-derived cells. In this study, we used Rac2-null (Rac2(-/-)) and Nox2(-/-) mice with a novel method of identifying primary hindlimb collaterals to investigate the hypothesis that collateral growth requires these molecules. When initial experiments performed with femoral ligation demonstrated similar perfusion and collateral growth in Rac2(-/-) and wild-type C57BL/6J (BL6) mice, subsequent experiments were performed with a more severe ischemia model, femoral artery excision. After femoral excision, tissue perfusion was suppressed in Rac2(-/-) mice relative to BL6 mice. Histological assessment of ischemic injury including necrotic and regenerated muscle fibers and lipid and collagen deposition demonstrated greater injury in Rac2(-/-) mice. The diameters of primary collaterals identified during Microfil injection with intravital microscopy were enlarged to a similar extent in BL6 and Rac2(-/-) mice. Intimal cells in collateral cross sections were increased in number in both strains and were CD31 positive and CD45 negative. Circulating leukocytes and CD11b(+) cells were increased more in Rac2(-/-) than BL6 animals. Experiments performed in Nox2(-/-) mice to verify that the unexpected results related to collateral growth were not unique to Rac2(-/-) mice gave equivalent results. The data demonstrate that, subsequent to acute femoral artery excision, perfusion recovery is impaired in Rac2(-/-) and Nox2(-/-) mice but that collateral luminal expansion and intimal cell recruitment/proliferation are normal. These novel results indicate that collateral luminal expansion and intimal cell recruitment/proliferation are not mediated by Rac2 and Nox2.


Assuntos
Circulação Colateral , Isquemia/fisiopatologia , Glicoproteínas de Membrana/deficiência , Músculo Esquelético/irrigação sanguínea , NADPH Oxidases/deficiência , Proteínas rac de Ligação ao GTP/deficiência , Animais , Células da Medula Óssea/patologia , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Artéria Femoral/cirurgia , Membro Posterior , Isquemia/enzimologia , Isquemia/patologia , Leucócitos/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/patologia , NADPH Oxidase 2 , NADPH Oxidases/genética , Neovascularização Fisiológica , Fluxo Sanguíneo Regional , Fatores de Tempo , Túnica Íntima/patologia , Proteínas rac de Ligação ao GTP/genética , Proteína RAC2 de Ligação ao GTP
16.
Antioxid Redox Signal ; 10(11): 1849-52, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18665798

RESUMO

The field of stem and progenitor cell biology is expanding. Much of the enthusiasm is based on the potential of using stem and progenitor cells as a cellular therapy for the treatment of human disease. Although the concept of using human embryonic stem cells for therapeutic indications is intriguing, significant challenges face investigators pursuing research in this area. Therefore, renewed scientific energy is focusing on the molecular pathways that differentiate a pluripotent embryonic stem cell from more-committed tissue-specific cells. Molecular mechanisms that govern tissue-specific stem and progenitor cell function are also topics of intense investigation, given that altered function of these cells may promote a variety of human pathologies including aging, vascular disease, and cancer. Considerable progress has been made, but a clear identification of the molecular signatures of stem and progenitor cells remains elusive. A growing body of literature demonstrates that distinct functional characteristics of stem and progenitor cells are under redox regulation. In this Forum Issue, evidence for redox regulation of tissue-specific stem and progenitor cells involved in hematopoiesis and vasculogenesis/angiogenesis is presented.


Assuntos
Transdução de Sinais , Células-Tronco/metabolismo , Animais , Hematopoese/fisiologia , Humanos , Oxirredução , Estresse Oxidativo/fisiologia , Células-Tronco/citologia , Doenças Vasculares/metabolismo , Doenças Vasculares/fisiopatologia
17.
Pediatr Res ; 64(1): 68-73, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18360305

RESUMO

Endothelial progenitor cells (EPCs) are used for angiogenic therapies and as biomarkers of cardiovascular disease. Human umbilical cord blood (UCB) is a rich source of endothelial colony forming cells (ECFCs), which are EPCs with robust proliferative potential that may be useful for clinical vascular regeneration. Previous studies show that hematopoietic progenitor cells are increased in premature UCB compared with term controls. Based on this paradigm, we hypothesized that premature UCB would be an enriched source of ECFCs. Thirty-nine UCB samples were obtained from premature infants (24-37 wk gestational age (GA)) and term controls. ECFC colonies were enumerated, clonally isolated, and identified by expression of endothelial cell surface antigens and functional analysis. GA of 33-36 wk UCB yielded predominantly ECFC colonies at equivalent numbers to term infants. UCB from 24 to 28 wk GA infants had significantly fewer ECFCs. Surprisingly, 24-28 wk GA UCB yielded predominantly mesenchymal stem cell (MSC) colonies, capable of differentiating into adipocytes, chondrocytes, and osteocytes. MSCs were rarely identified in 37-40 wk GA UCB. These studies demonstrate that circulating MSCs and ECFCs appear at different GA in the human UCB, and that 24-28 wk GA UCB may be a novel source of MSCs for therapeutic use in human diseases.


Assuntos
Células Endoteliais/fisiologia , Sangue Fetal/citologia , Células-Tronco Fetais/fisiologia , Células-Tronco Mesenquimais/fisiologia , Adipócitos/fisiologia , Antígenos CD/análise , Diferenciação Celular , Proliferação de Células , Forma Celular , Células Cultivadas , Condrócitos/fisiologia , Células Endoteliais/imunologia , Células-Tronco Fetais/imunologia , Idade Gestacional , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Células-Tronco Mesenquimais/imunologia , Neovascularização Fisiológica , Osteócitos/fisiologia , Fenótipo
18.
Diabetes ; 57(3): 724-31, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18086900

RESUMO

OBJECTIVE: Emerging data demonstrate that maternal diabetes has long-term health consequences for offspring, including the development of hypertension. In adults, circulating endothelial progenitor cells (EPCs) participate in vascular repair, and EPC numbers and function inversely correlate with the risk of developing vascular disease. Therefore, our objectives were to determine whether hyperglycemia or exposure to a diabetic intrauterine environment alters EPC function. RESEARCH DESIGN AND METHODS: We used well-established clonogenic endothelial colony-forming cell (ECFC) assays and murine transplantation experiments to examine human vasculogenesis. RESULTS: Both in vitro hyperglycemia and a diabetic intrauterine environment reduced ECFC colony formation, self-renewal capacity, and capillary-like tube formation in matrigel. This cellular phenotype was linked to premature senescence and reduced proliferation. Further, cord blood ECFCs from diabetic pregnancies formed fewer chimeric vessels de novo after transplantation into immunodeficient mice compared with neonatal ECFCs harvested from uncomplicated pregnancies. CONCLUSIONS; Collectively, these data demonstrate that hyperglycemia or exposure to a diabetic intrauterine environment diminishes neonatal ECFC function both in vitro and in vivo, providing potential mechanistic insights into the long-term cardiovascular complications observed in newborns of diabetic pregnancies.


Assuntos
Diabetes Gestacional , Células Endoteliais/efeitos dos fármacos , Hiperglicemia , Células-Tronco/efeitos dos fármacos , Adulto , Proliferação de Células , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Glucose/farmacologia , Humanos , Recém-Nascido , Gravidez
19.
Blood ; 110(7): 2704-7, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17616641

RESUMO

Several hematopoietic stem-cell (HSC) regulators are controlled by ubiquitin-mediated proteolysis, so the ubiquitin pathway might modulate HSC function. However, this hypothesis has not been formally tested. Cul4A encodes a core subunit of one ubiquitin ligase. Whereas Cul4A-deficient embryos die in utero, Cul4A-haploinsufficient mice are viable but exhibit abnormal hematopoiesis (fewer erythroid and primitive myeloid progenitors). Given these data, we examined whether Cul4A(+/-) HSCs might also be impaired. Using bone marrow transplantation assays, we determined that Cul4A(+/-) HSCs exhibit defects in engraftment and self-renewal capacity. These studies are the first to demonstrate that ubiquitin-mediated protein degradation is important for HSC function. Further, they indicate that a Cul4A ubiquitin ligase targets for degradation one or multiple HSC regulators.


Assuntos
Proteínas Culina/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Proliferação de Células , Quimerismo , Proteínas Culina/genética , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Camundongos , Camundongos Transgênicos
20.
Exp Hematol ; 35(7): 1109-18, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17588480

RESUMO

OBJECTIVE: Endothelial progenitor cells (EPCs) are used for angiogenic therapies or as biomarkers to assess cardiovascular disease risk. However, there is no uniform definition of an EPC, which confounds EPC studies. EPCs are widely described as cells that coexpress the cell-surface antigens CD34, AC133, and vascular endothelial growth factor receptor-2 (VEGFR-2). These antigens are also expressed on primitive hematopoietic progenitor cells (HPCs). Remarkably, despite their original identification, CD34+AC133+VEGFR-2+ cells have never been isolated and simultaneously plated in hematopoietic and endothelial cell (EC) clonogenic assays to assess the identity of their clonal progeny, which are presumably the cellular participants in vascular regeneration. METHODS: CD34+AC133+VEGFR-2+ cells were isolated from human umbilical cord blood (CB) or granulocyte colony-stimulating factor-mobilized peripheral blood and assayed for either EPCs or HPCs. RESULTS: CD34+AC133+VEGFR-2+ cells did not form EPCs and were devoid of vessel forming activity. However, CD34+AC133+VEGFR-2+ cells formed HPCs and expressed the hematopoietic lineage-specific antigen, CD45. We next tested whether EPCs could be separated from HPCs by immunoselection for CD34 and CD45. CD34+CD45+ cells formed HPCs but not EPCs, while CD34+CD45- cells formed EPCs but not HPCs. CONCLUSIONS: Therefore, CD34+AC133+VEGFR-2+ cells are HPCs that do not yield EC progeny, and the biological mechanism for their correlation with cardiovascular disease needs to be reexamined.


Assuntos
Antígenos CD34/análise , Antígenos CD/análise , Células Endoteliais/citologia , Glicoproteínas/análise , Células-Tronco Hematopoéticas/citologia , Peptídeos/análise , Células-Tronco/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Antígeno AC133 , Adulto , Separação Celular , Células Cultivadas , Feminino , Humanos , Antígenos Comuns de Leucócito/análise , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA