Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 42(1): e1-e9, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34758632

RESUMO

OBJECTIVE: Antibody blockade of the "do not eat me" signal CD47 (cluster of differentiation 47) enhances efferocytosis and reduces lesion size and necrotic core formation in murine atherosclerosis. TNF (Tumor necrosis factor)-α expression directly enhances CD47 expression, and elevated TNF-α is observed in the absence of the proefferocytosis receptor LRP1 (low-density lipoprotein receptor-related protein 1), a regulator of atherogenesis and inflammation. Thus, we tested the hypothesis that CD47 blockade requires the presence of macrophage LRP1 to enhance efferocytosis, temper TNF-α-dependent inflammation, and limit atherosclerosis. Approach and Results: Mice lacking systemic apoE (apoE-/-), alone or in combination with the loss of macrophage LRP1 (double knockout), were fed a Western-type diet for 12 weeks while receiving anti-CD47 antibody (anti-CD47) or IgG every other day. In apoE-/- mice, treatment with anti-CD47 reduced lesion size by 25.4%, decreased necrotic core area by 34.5%, and decreased the ratio of free:macrophage-associated apoptotic bodies by 47.6% compared with IgG controls (P<0.05), confirming previous reports. Double knockout mice treated with anti-CD47 showed no differences in lesion size, necrotic core area, or the ratio of free:macrophage-associated apoptotic bodies compared with IgG controls. In vitro efferocytosis was 30% higher when apoE-/- phagocytes were incubated with anti-CD47 compared with IgG controls (P<0.05); however, anti-CD47 had no effect on efferocytosis in double knockout phagocytes. Analyses of mRNA and protein showed increased CD47 expression in anti-inflammatory IL (interleukin)-4 treated LRP1-/- macrophages compared with wild type, but no differences were observed in inflammatory lipopolysaccharide-treated macrophages. CONCLUSIONS: The proefferocytosis receptor LRP1 in macrophages is necessary for anti-CD47 blockade to enhance efferocytosis, limit atherogenesis, and decrease necrotic core formation in the apoE-/- model of atherosclerosis.


Assuntos
Anti-Inflamatórios/farmacologia , Anticorpos Bloqueadores/farmacologia , Aorta/efeitos dos fármacos , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Antígeno CD47/antagonistas & inibidores , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Macrófagos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Animais , Aorta/imunologia , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/imunologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Antígeno CD47/imunologia , Antígeno CD47/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Necrose , Placa Aterosclerótica , Fator de Necrose Tumoral alfa/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(27): 15818-15826, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32541024

RESUMO

Atherosclerosis is the process underlying heart attack and stroke. Despite decades of research, its pathogenesis remains unclear. Dogma suggests that atherosclerotic plaques expand primarily via the accumulation of cholesterol and inflammatory cells. However, recent evidence suggests that a substantial portion of the plaque may arise from a subset of "dedifferentiated" vascular smooth muscle cells (SMCs) which proliferate in a clonal fashion. Herein we use multicolor lineage-tracing models to confirm that the mature SMC can give rise to a hyperproliferative cell which appears to promote inflammation via elaboration of complement-dependent anaphylatoxins. Despite being extensively opsonized with prophagocytic complement fragments, we find that this cell also escapes immune surveillance by neighboring macrophages, thereby exacerbating its relative survival advantage. Mechanistic studies indicate this phenomenon results from a generalized opsonin-sensing defect acquired by macrophages during polarization. This defect coincides with the noncanonical up-regulation of so-called don't eat me molecules on inflamed phagocytes, which reduces their capacity for programmed cell removal (PrCR). Knockdown or knockout of the key antiphagocytic molecule CD47 restores the ability of macrophages to sense and clear opsonized targets in vitro, allowing for potent and targeted suppression of clonal SMC expansion in the plaque in vivo. Because integrated clinical and genomic analyses indicate that similar pathways are active in humans with cardiovascular disease, these studies suggest that the clonally expanding SMC may represent a translational target for treating atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Clonagem Molecular , Ativação do Complemento , Miócitos de Músculo Liso/metabolismo , Fagocitose/fisiologia , Animais , Antígeno CD47/metabolismo , Linhagem da Célula , Proliferação de Células , Complemento C3/genética , Complemento C3/metabolismo , Feminino , Humanos , Inflamação , Macrófagos/metabolismo , Masculino , Camundongos Knockout para ApoE , Miócitos de Músculo Liso/citologia , Placa Aterosclerótica/metabolismo , Análise de Sequência de RNA , Regulação para Cima
3.
Nature ; 536(7614): 86-90, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27437576

RESUMO

Atherosclerosis is the disease process that underlies heart attack and stroke. Advanced lesions at risk of rupture are characterized by the pathological accumulation of diseased vascular cells and apoptotic cellular debris. Why these cells are not cleared remains unknown. Here we show that atherogenesis is associated with upregulation of CD47, a key anti-phagocytic molecule that is known to render malignant cells resistant to programmed cell removal, or 'efferocytosis'. We find that administration of CD47-blocking antibodies reverses this defect in efferocytosis, normalizes the clearance of diseased vascular tissue, and ameliorates atherosclerosis in multiple mouse models. Mechanistic studies implicate the pro-atherosclerotic factor TNF-α as a fundamental driver of impaired programmed cell removal, explaining why this process is compromised in vascular disease. Similar to recent observations in cancer, impaired efferocytosis appears to play a pathogenic role in cardiovascular disease, but is not a fixed defect and may represent a novel therapeutic target.


Assuntos
Anticorpos Bloqueadores/imunologia , Anticorpos Bloqueadores/farmacologia , Aterosclerose/prevenção & controle , Antígeno CD47/imunologia , Fagocitose/efeitos dos fármacos , Animais , Apoptose , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/terapia , Antígeno CD47/biossíntese , Antígeno CD47/metabolismo , Artérias Carótidas/patologia , Vasos Coronários/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , NF-kappa B/metabolismo , Biossíntese de Proteínas , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
4.
Arterioscler Thromb Vasc Biol ; 40(12): 2821-2828, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33086865

RESUMO

OBJECTIVE: This study sought to determine whether 18F-fluorodeoxyglucose-positron emission tomography/computed tomography could be applied to a murine model of advanced atherosclerotic plaque vulnerability to detect response to therapeutic intervention and changes in lesion stability. Approach and Results: To analyze plaques susceptible to rupture, we fed ApoE-/- mice a high-fat diet and induced vulnerable lesions by cast placement over the carotid artery. After 9 weeks of treatment with orthogonal therapeutic agents (including lipid-lowering and proefferocytic therapies), we assessed vascular inflammation and several features of plaque vulnerability by 18F-fluorodeoxyglucose-positron emission tomography/computed tomography and histopathology, respectively. We observed that 18F-fluorodeoxyglucose-positron emission tomography/computed tomography had the capacity to resolve histopathologically proven changes in plaque stability after treatment. Moreover, mean target-to-background ratios correlated with multiple characteristics of lesion instability, including the corrected vulnerability index. CONCLUSIONS: These results suggest that the application of noninvasive 18F-fluorodeoxyglucose-positron emission tomography/computed tomography to a murine model can allow for the identification of vulnerable atherosclerotic plaques and their response to therapeutic intervention. This approach may prove useful as a drug discovery and prioritization method.


Assuntos
Doenças das Artérias Carótidas/diagnóstico por imagem , Artéria Carótida Primitiva/diagnóstico por imagem , Fluordesoxiglucose F18/administração & dosagem , Placa Aterosclerótica , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos/administração & dosagem , Animais , Anticorpos Bloqueadores/farmacologia , Atorvastatina/farmacologia , Antígeno CD47/antagonistas & inibidores , Doenças das Artérias Carótidas/tratamento farmacológico , Doenças das Artérias Carótidas/patologia , Artéria Carótida Primitiva/efeitos dos fármacos , Artéria Carótida Primitiva/patologia , Modelos Animais de Doenças , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Valor Preditivo dos Testes , Ruptura Espontânea
5.
Arterioscler Thromb Vasc Biol ; 39(4): 635-646, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30786744

RESUMO

Nanoparticles promise to advance strategies to treat vascular disease. Since being harnessed by the cancer field to deliver safer and more effective chemotherapeutics, nanoparticles have been translated into applications for cardiovascular disease. Systemic exposure and drug-drug interactions remain a concern for nearly all cardiovascular therapies, including statins, antithrombotic, and thrombolytic agents. Moreover, off-target effects and poor bioavailability have limited the development of completely new approaches to treat vascular disease. Through the rational design of nanoparticles, nano-based delivery systems enable more efficient delivery of a drug to its therapeutic target or even directly to the diseased site, overcoming biological barriers and enhancing a drug's therapeutic index. In addition, advances in molecular imaging have led to the development of theranostic nanoparticles that may simultaneously act as carriers of both therapeutic and imaging payloads. The following is a summary of nanoparticle therapy for atherosclerosis, thrombosis, and restenosis and an overview of recent major advances in the targeted treatment of vascular disease.


Assuntos
Fármacos Cardiovasculares/administração & dosagem , Portadores de Fármacos/administração & dosagem , Nanopartículas/administração & dosagem , Doenças Vasculares/tratamento farmacológico , Animais , Quimiotaxia de Leucócito/efeitos dos fármacos , Colesterol/metabolismo , Avaliação Pré-Clínica de Medicamentos , Previsões , Humanos , Inflamação/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Camundongos , Neointima/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/fisiopatologia , Placa Aterosclerótica/fisiopatologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Trombose/tratamento farmacológico
6.
Circ Res ; 118(2): 230-40, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26596284

RESUMO

RATIONALE: Genetic variation at the chromosome 9p21 cardiovascular risk locus has been associated with peripheral artery disease, but its mechanism remains unknown. OBJECTIVE: To determine whether this association is secondary to an increase in atherosclerosis, or it is the result of a separate angiogenesis-related mechanism. METHODS AND RESULTS: Quantitative evaluation of human vascular samples revealed that carriers of the 9p21 risk allele possess a significantly higher burden of immature intraplaque microvessels than carriers of the ancestral allele, irrespective of lesion size or patient comorbidity. To determine whether aberrant angiogenesis also occurs under nonatherosclerotic conditions, we performed femoral artery ligation surgery in mice lacking the 9p21 candidate gene, Cdkn2b. These animals developed advanced hindlimb ischemia and digital autoamputation, secondary to a defect in the capacity of the Cdkn2b-deficient smooth muscle cell to support the developing neovessel. Microarray studies identified impaired transforming growth factor ß (TGFß) signaling in cultured cyclin-dependent kinase inhibitor 2B (CDKN2B)-deficient cells, as well as TGFß1 upregulation in the vasculature of 9p21 risk allele carriers. Molecular signaling studies indicated that loss of CDKN2B impairs the expression of the inhibitory factor, SMAD-7, which promotes downstream TGFß activation. Ultimately, this manifests in the upregulation of a poorly studied effector molecule, TGFß1-induced-1, which is a TGFß-rheostat known to have antagonistic effects on the endothelial cell and smooth muscle cell. Dual knockdown studies confirmed the reversibility of the proposed mechanism, in vitro. CONCLUSIONS: These results suggest that loss of CDKN2B may not only promote cardiovascular disease through the development of atherosclerosis but may also impair TGFß signaling and hypoxic neovessel maturation.


Assuntos
Aterosclerose/enzimologia , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Neovascularização Fisiológica , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/mortalidade , Aterosclerose/patologia , Artérias Carótidas/enzimologia , Artérias Carótidas/patologia , Hipóxia Celular , Células Cultivadas , Cromossomos Humanos Par 9 , Vasos Coronários/enzimologia , Vasos Coronários/patologia , Inibidor de Quinase Dependente de Ciclina p15/deficiência , Inibidor de Quinase Dependente de Ciclina p15/genética , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Membro Posterior , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/fisiopatologia , Neovascularização Patológica , Fenótipo , Interferência de RNA , Proteína Smad7/metabolismo , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta1/genética
8.
Circulation ; 131(13): 1160-70, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25825396

RESUMO

BACKGROUND: Pattern recognition receptor nucleotide-binding oligomerization domain 2 (NOD2) is well investigated in immunity, but its expression and function in platelets has never been explored. METHOD AND RESULTS: Using reverse transcription polymerase chain reaction and Western blot, we show that both human and mouse platelets express NOD2, and its agonist muramyl dipeptide induced NOD2 activation as evidenced by receptor dimerization. NOD2 activation potentiates platelet aggregation and secretion induced by low concentrations of thrombin or collagen, and clot retraction, as well. These potentiating effects of muramyl dipeptide were not seen in platelets from NOD2-deficient mice. Plasma from septic patients also potentiates platelet aggregation induced by thrombin or collagen NOD2 dependently. Using intravital microscopy, we found that muramyl dipeptide administration accelerated in vivo thrombosis in a FeCl3-injured mesenteric arteriole thrombosis mouse model. Platelet depletion and transfusion experiments confirmed that NOD2 from platelets contributes to the in vivo thrombosis in mice. NOD2 activation also accelerates platelet-dependent hemostasis. We further found that platelets express receptor-interacting protein 2, and provided evidence suggesting that mitogen activated-protein kinase and nitric oxide/soluble guanylyl cyclase/cGMP/protein kinase G pathways downstream of receptor-interacting protein mediate the role of NOD2 in platelets. Finally, muramyl dipeptide stimulates proinflammatory cytokine interleukin-1ß maturation and accumulation in human and mouse platelets NOD2 dependently. CONCLUSIONS: NOD2 is expressed in platelets and functions in platelet activation and arterial thrombosis, possibly during infection. To our knowledge, this is the first study on NOD-like receptors in platelets that link thrombotic events to inflammation.


Assuntos
Plaquetas/metabolismo , Inflamação/sangue , Proteína Adaptadora de Sinalização NOD2/fisiologia , Ativação Plaquetária/fisiologia , Trombose/sangue , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Animais , Bacteriemia/sangue , Plaquetas/efeitos dos fármacos , Retração do Coágulo/fisiologia , GMP Cíclico/sangue , Dimerização , Hemostasia/fisiologia , Humanos , Interleucina-1beta/sangue , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Óxido Nítrico/sangue , Proteína Adaptadora de Sinalização NOD2/agonistas , Proteína Adaptadora de Sinalização NOD2/biossíntese , Proteína Adaptadora de Sinalização NOD2/sangue , Ativação Plaquetária/efeitos dos fármacos , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/biossíntese , Proteína Serina-Treonina Quinases de Interação com Receptores/biossíntese , Transdução de Sinais/fisiologia
9.
Heart Fail Rev ; 20(5): 613-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26024953

RESUMO

Injection of various stem cells has been tested with the hopes of improving cardiac function after a myocardial infarction (MI). However, there is continued controversy as to which cell type is best for repair. Due to technical differences in cell isolation, processing, delivery, and cardiac functional assessment by various investigators, it has been difficult to directly compare the results of different cells. Using same techniques to evaluate the efficacy of different cell types, we have separately delivered bone marrow cells (BMCs), cardiospheres (CSs), CS-derived Sca-1(+)/CD45(-) cells, human embryonic stem cell-derived cardiomyocytes, and BMC extract into infarcted murine myocardium and found that all of these treatments reduce infarct size and improve cardiac function post-MI similarly without one regimen being superior to another. The beneficial effects appear to be via paracrine influences. Different progenitors lead to improved cardiac function post-MI, but it is premature to hype any specific cell type at this time.


Assuntos
Transplante de Medula Óssea/métodos , Infarto do Miocárdio , Miócitos Cardíacos/fisiologia , Transplante de Células-Tronco/métodos , Células-Tronco , Animais , Humanos , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/terapia , Células-Tronco/classificação , Células-Tronco/metabolismo , Resultado do Tratamento
10.
Platelets ; 26(8): 736-44, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25734213

RESUMO

Aberrant platelet activation plays a critical role in the pathogenesis of heart attack and stroke. DL-3-n-butylphthalide (NBP) has been approved in China to treat stroke with multiple mechanisms. The anti-stroke effects of NBP may be related to its antiplatelet effects reported in rats in addition to its antioxidative, antiapoptotic, and angiogenic effects. However, the effects and the underlying mechanisms of NBP on human platelets are not yet clear. In this study, we found that NBP concentration-dependently inhibited human platelet aggregation and ATP release induced by ADP, thrombin, U46619, arachidonic acid, or collagen. NBP also inhibited PAC-1 binding induced by ADP or thrombin and platelet spreading on immobilized fibrinogen. NBP reduced TXA2 synthesis induced by thrombin or collagen via inhibiting cPLA2 phosphorylation, concomitantly with a marked decrease in intracellular calcium mobilization. Moreover, NBP also inhibited human platelet phosphodiesterase (PDE) and elevated 3,5-cyclic adenosine monophosphate level in platelets. In conclusion, NBP significantly inhibits human platelet activation via inhibition of cPLA2-mediated TXA2 synthesis and PDE, and may be effective as an antiplatelet drug to treat other arterial thrombotic diseases.


Assuntos
Benzofuranos/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Fosfolipases A2 Citosólicas/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Tromboxano A2/biossíntese , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/metabolismo , Difosfato de Adenosina/farmacologia , Cálcio/metabolismo , Colágeno/farmacologia , AMP Cíclico/metabolismo , Humanos , Fosforilação , Trombina/farmacologia
12.
J Cardiovasc Pharmacol ; 63(2): 85-94, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23674056

RESUMO

Various stem cell types have been tested for regenerating damaged myocardium after myocardial infarction. However, the results of clinical trials have not been consistent, with only some of the trials reporting small improvements in cardiac function. It seems that engraftment and survival of injected cells is limited and transplanted stem cells either do not differentiate into cardiac cells or differentiate into only limited number of cardiac cells. The exact mechanism(s) of cardiac functional improvement by cell therapy are unclear, but paracrine effect may play a central role. The resident cardiac progenitor cells identified within the adult myocardium have distinct advantages over other stem cell types for cardiac cell therapy, as they are likely precommitted to the cardiovascular fate. However, isolating and expanding these cells from cardiac biopsies is a challenge. More recently, direct reprogramming of fibroblasts into cardiomyocytes has given new hope for myocardial regeneration. Here we will review different stem cells used in cardiac cell therapy with a focus on the native cardiac progenitor cells and briefly outline future directions of cardiac cell therapy.


Assuntos
Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Adulto , Animais , Diferenciação Celular/fisiologia , Ensaios Clínicos como Assunto , Fibroblastos/citologia , Humanos , Infarto do Miocárdio/fisiopatologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Regeneração , Transplante de Células-Tronco/tendências
13.
J Cardiovasc Pharmacol ; 63(5): 406-11, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24805144

RESUMO

Cardiovascular disease is the leading cause of death in Western countries. A major limitation of current treatments is the inability to efficiently repair or replace dead myocardium. Recently, stem cell-based therapies have been explored as an avenue to circumvent current therapeutic limitations. Overall, these therapies seem to result in small improvements in the contractile function of the heart. The exact mechanism(s) of action that underlie these improvements remain unknown, and it is believed that paracrine effects play a significant role. Previously, we had reported that an extract derived from bone marrow cells, in the absence of any live cell, contained cardioprotective soluble factors. In this study, we identify IL-15 as a putative cardioprotectant within the bone marrow cells paracrine profile. Using an in vitro culture system, we assessed the ability of IL-15 to protect cardiomyocytes under hypoxic conditions. For the first time, we have identified IL-15 receptors on the surface of cardiomyocytes and delineated the signaling system by which hypoxic cardiomyocytes may be protected from cellular death and rescued from oxidative stress with IL-15 treatment.


Assuntos
Interleucina-15/fisiologia , Miócitos Cardíacos/fisiologia , Transdução de Sinais/fisiologia , Animais , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Receptores de Interleucina-15/efeitos dos fármacos , Receptores de Interleucina-15/metabolismo , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/efeitos dos fármacos
14.
Patient Prefer Adherence ; 18: 1095-1105, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38854479

RESUMO

Purpose: Stroke is a major disease endangering the health of Chinese people, and patients need to rely on the care of family members, which brings heavy caregiving burdens and pressures to caregivers and families, thus disrupting the stable family structure. In view of this, this study was to analyse the current status of family resilience among caregivers of stroke patients in Chinese nuclear families, and to explore the correlation and mechanism of action among perceived stress, illness uncertainty and family resilience. Patients and Methods: This study used a cross-sectional research design. A total of 350 carers of stroke patients in nuclear families from four tertiary hospitals in Suzhou City, Jiangsu Province, China were selected by convenience sampling method and assessed by using demographic questionnaires, the Chinese Perceived Stress Scale (CPSS), the Parental Perceptions of Uncertainty Scale-Family (PPUS-FM), and a short Chinese version of the Family Resilience Assessment Scale (FRAS-C). Based on the above data, structural equation model was used to test the mediating role of perceived stress between illness uncertainty and family resilience. Results: Family resilience among caregivers of stroke patients in nuclear families was at the medium lower level, illness uncertainty was at the medium level, and perceived stress was at the relatively high level. Illness uncertainty was positively correlated with perceived stress (P<0.01) and negatively correlated with family resilience (P<0.01). Illness uncertainty directly predicted family resilience (ß = -0.516, p < 0.05). And the pathway between illness uncertainty and family resilience was partially mediated by perceived stress (Effect= -0.091, 95% CI [-0.141, -0.055]). Conclusion: Healthcare professionals should pay adequate attention to the level of illness uncertainty and perceived stress among carers of stroke patients, with the need to take measures to reduce carers' illness uncertainty and perceived stress in order to improve family resilience.

15.
PLoS One ; 19(5): e0303610, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38758931

RESUMO

We have previously shown that polygenic risk scores (PRS) can improve risk stratification of peripheral artery disease (PAD) in a large, retrospective cohort. Here, we evaluate the potential of PRS in improving the detection of PAD and prediction of major adverse cardiovascular and cerebrovascular events (MACCE) and adverse events (AE) in an institutional patient cohort. We created a cohort of 278 patients (52 cases and 226 controls) and fit a PAD-specific PRS based on the weighted sum of risk alleles. We built traditional clinical risk models and machine learning (ML) models using clinical and genetic variables to detect PAD, MACCE, and AE. The models' performances were measured using the area under the curve (AUC), net reclassification index (NRI), integrated discrimination improvement (IDI), and Brier score. We also evaluated the clinical utility of our PAD model using decision curve analysis (DCA). We found a modest, but not statistically significant improvement in the PAD detection model's performance with the inclusion of PRS from 0.902 (95% CI: 0.846-0.957) (clinical variables only) to 0.909 (95% CI: 0.856-0.961) (clinical variables with PRS). The PRS inclusion significantly improved risk re-classification of PAD with an NRI of 0.07 (95% CI: 0.002-0.137), p = 0.04. For our ML model predicting MACCE, the addition of PRS did not significantly improve the AUC, however, NRI analysis demonstrated significant improvement in risk re-classification (p = 2e-05). Decision curve analysis showed higher net benefit of our combined PRS-clinical model across all thresholds of PAD detection. Including PRS to a clinical PAD-risk model was associated with improvement in risk stratification and clinical utility, although we did not see a significant change in AUC. This result underscores the potential clinical utility of incorporating PRS data into clinical risk models for prevalent PAD and the need for use of evaluation metrics that can discern the clinical impact of using new biomarkers in smaller populations.


Assuntos
Doença Arterial Periférica , Humanos , Doença Arterial Periférica/genética , Doença Arterial Periférica/diagnóstico , Feminino , Masculino , Idoso , Pessoa de Meia-Idade , Medição de Risco/métodos , Fatores de Risco , Aprendizado de Máquina , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/diagnóstico , Estudos Retrospectivos , Herança Multifatorial/genética , Estudos de Casos e Controles , Área Sob a Curva , Estratificação de Risco Genético
16.
Cell Tissue Res ; 351(1): 201-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23064904

RESUMO

The cardiosphere (CS) is composed of a heterogeneous population of cells, including CD45(+) cells that are bone marrow (BM)-derived. However, whether the CD45(+) cells are an essential cell component in CS formation is unknown. The current study was undertaken to address this question. Cardiospheres (CSs) were harvested from 1-week post-myocardial infarction (MI) or non-MI hearts of C57BL/6 J mice. The process of CS formation was observed by timelapse photography. To analyze the role of BM-derived CD45(+) cells in CS formation, CD45(+) cells were depleted from populations of CS-forming cells by immunomagnetic beads. We recorded the number of CSs formed in culture from the same amount (10(5)) of intact CS-forming cells, from CD45(+)-cell-depleted CS-forming cells and from CD45(+) cells alone (n=6-9/cell type). CS-forming cells selectively aggregated together to form CSs by 35 h after plating. The depletion of CD45(+) cells from CS-forming cells actually increased the formation of CSs (67±10 CSs/10(5) cells) compared with non-depleted CS-forming cells (51±6 CSs/10(5) cells, P<0.0001). Purified CD45(+) cells from CS-forming cells did not form CSs in culture. Thus, BM-derived CD45(+) cells including BM progenitors are neither necessary nor sufficient for CS formation.


Assuntos
Antígenos Comuns de Leucócito/metabolismo , Miócitos Cardíacos/citologia , Esferoides Celulares/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Proliferação de Células , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Esferoides Celulares/metabolismo
17.
Nat Cardiovasc Res ; 1(3): 253-262, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35990913

RESUMO

The pleiotropic benefits of statins may result from their impact on vascular inflammation. The molecular process underlying this phenomenon is not fully elucidated. Here, RNA sequencing designed to investigate gene expression patterns following CD47-SIRPα inhibition identifies a link between statins, efferocytosis, and vascular inflammation. In vivo and in vitro studies provide evidence that statins augment programmed cell removal by inhibiting the nuclear translocation of NFκB1 p50 and suppressing the expression of the critical 'don't eat me' molecule, CD47. Statins amplify the phagocytic capacity of macrophages, and thus the anti-atherosclerotic effects of CD47-SIRPα blockade, in an additive manner. Analyses of clinical biobank specimens suggest a similar link between statins and CD47 expression in humans, highlighting the potential translational implications. Taken together, our findings identify efferocytosis and CD47 as pivotal mediators of statin pleiotropy. In turn, statins amplify the anti-atherosclerotic effects of pro-phagocytic therapies independently of any lipid-lowering effect.

18.
Cardiovasc Res ; 118(13): 2792-2804, 2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34849613

RESUMO

AIMS: De-differentiation and activation of pro-inflammatory pathways are key transitions vascular smooth muscle cells (SMCs) make during atherogenesis. Here, we explored the upstream regulators of this 'atherogenic transition'. METHODS AND RESULTS: Genome-wide sequencing studies, including Assay for Transposase-Accessible Chromatin using sequencing and RNA-seq, were performed on cells isolated from both murine SMC-lineage-tracing models of atherosclerosis and human atherosclerotic lesions. At the bulk level, alterations in chromatin accessibility were associated with the atherogenic transitioning of lesional SMCs, especially in relation to genes that govern differentiation status and complement-dependent inflammation. Using computational biology, we observed that a transcription factor previously related to coronary artery disease, Activating transcription factor 3 (ATF3), was predicted to be an upstream regulator of genes altered during the transition. At the single-cell level, our results indicated that ATF3 is a key repressor of SMC transitioning towards the subset of cells that promote vascular inflammation by activating the complement cascade. The expression of ATF3 and complement component C3 was negatively correlated in SMCs from human atherosclerotic lesions, suggesting translational relevance. Phenome-wide association studies indicated that genetic variation that results in reduced expression of ATF3 is correlated with an increased risk for atherosclerosis, and the expression of ATF3 was significantly down-regulated in humans with advanced vascular disease. CONCLUSION: Our study indicates that the plasticity of atherosclerotic SMCs may in part be explained by dynamic changes in their chromatin architecture, which in turn may contribute to their maladaptive response to inflammation-induced stress.


Assuntos
Aterosclerose , Músculo Liso Vascular , Humanos , Camundongos , Animais , Músculo Liso Vascular/metabolismo , Cromatina/genética , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Miócitos de Músculo Liso/metabolismo , Aterosclerose/metabolismo , Inflamação/metabolismo
19.
J Transl Med ; 9: 150, 2011 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-21910857

RESUMO

BACKGROUND: Stem cell homing to the heart is mediated by the release of chemo-attractant cytokines. Stromal derived factor -1 alpha (SDF-1a) and monocyte chemotactic factor 1(MCP-1) are detectable in peripheral blood after myocardial infarction (MI). It remains unknown if they are produced by, and released from, the heart in order to attract stem cells to repair the damaged myocardium. METHODS: Murine hearts were studied for expression of MCP-1 and SDF-1a at day 3 and day 28 following myocardial infarction to determine whether production is increased following MI. In addition, we studied the coronary artery and coronary sinus (venous) blood from patients with normal coronary arteries, stable coronary artery disease (CAD), unstable angina and MI to determine whether these cytokines are released from the heart into the systemic circulation following MI. RESULTS: Both MCP-1 and SDF-1a are constitutively produced and released by the heart. MCP-1 mRNA is upregulated following murine experimental MI, but SDF-1a is suppressed. There is less release of SDF-1a into the systemic circulation in patients with all stages of CAD including MI, mimicking the animal model. However MCP-1 release from the human heart following MI is also suppressed, which is the exact opposite of the animal model. CONCLUSIONS: SDF-1a and MCP-1 release from the human heart are suppressed following MI. In the case of SDF-1a, the animal model appropriately reflects the human situation. However, for MCP-1 the animal model is the exact opposite of the human condition. Human observational studies like this one are paramount in guiding translation from experimental studies to clinical trials.


Assuntos
Quimiocina CCL2/metabolismo , Quimiocina CXCL12/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Idoso , Animais , Quimiocina CCL2/genética , Quimiocina CXCL12/genética , Demografia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo
20.
Ann Neurol ; 67(4): 488-97, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20437584

RESUMO

OBJECTIVE: Endothelial progenitor cells (EPCs) play an important role in tissue repairing and regeneration in ischemic organs, including the brain. However, the cause of EPC migration and the function of EPCs after ischemia are unclear. In this study, we demonstrated the effects of EPCs on ischemic brain injury in a mouse model of transient middle cerebral artery occlusion (tMCAO). METHODS: Circulating human EPCs were characterized with immunofluorescent staining and flow cytometry. EPCs (1 x 10(6)) were injected into nude mice after 1 hour of tMCAO. Histological analysis and behavioral tests were performed from day 0 to 28 days after tMCAO. RESULTS: EPCs were detected in ischemic brain regions 24 hours after tMCAO. EPC transplantation significantly reduced ischemic infarct volume at 3 days after tMCAO compared with control animals (p < 0.05). CXCR4 was expressed in the majority of EPCs, and stromal-derived factor-1 (SDF-1) induced EPC migration, which was blocked by pretreated EPCs with AMD3100 in vitro. SDF-1 was upregulated in ischemic brain. Compared with control animals, injecting AMD3100-pretreated EPCs resulted in a larger infarct volume 3 days after tMCAO, suggesting that SDF-1-mediated signaling was involved in EPC-mediated neuroprotection. In addition, EPC transplantation reduced mouse cortex atrophy 4 weeks after tMCAO and improved neurobehavioral outcomes (p < 0.05). EPC injection potently increased angiogenesis in the peri-infarction area (p < 0.05). INTERPRETATION: We conclude that systemic delivery of EPCs protects the brain against ischemic injury, promotes neurovascular repair, and improves long-term neurobehavioral outcomes. Our data suggest that SDF-1-mediated signaling plays a critical role in EPC-mediated neuroprotection.


Assuntos
Células Endoteliais/transplante , Endotélio/citologia , Infarto da Artéria Cerebral Média/cirurgia , Transplante de Células-Tronco/métodos , Análise de Variância , Animais , Antígenos CD34/metabolismo , Comportamento Animal , Benzilaminas , Antígeno CD11b/metabolismo , Caderinas/metabolismo , Capilares/patologia , Infarto Cerebral/etiologia , Infarto Cerebral/prevenção & controle , Quimiocina CXCL12/metabolismo , Ciclamos , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Citometria de Fluxo/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Compostos Heterocíclicos/farmacologia , Humanos , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/fisiopatologia , Injeções Intravenosas/métodos , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Nus , Atividade Motora/fisiologia , Neovascularização Fisiológica/fisiologia , Desempenho Psicomotor , Receptores CXCR4/metabolismo , Fatores de Tempo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator de von Willebrand/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA