Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
bioRxiv ; 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38370768

RESUMO

To investigate the co-development of vasculature, mesenchyme, and epithelium crucial for organogenesis and the acquisition of organ-specific characteristics, we constructed a human pluripotent stem cell-derived organoid system comprising lung or intestinal epithelium surrounded by organotypic mesenchyme and vasculature. We demonstrated the pivotal role of co-differentiating mesoderm and endoderm via precise BMP regulation in generating multilineage organoids and gut tube patterning. Single-cell RNA-seq analysis revealed organ specificity in endothelium and mesenchyme, and uncovered key ligands driving endothelial specification in the lung (e.g., WNT2B and Semaphorins) or intestine (e.g., GDF15). Upon transplantation under the kidney capsule in mice, these organoids further matured and developed perfusable human-specific sub-epithelial capillaries. Additionally, our model recapitulated the abnormal endothelial-epithelial crosstalk in patients with FOXF1 deletion or mutations. Multilineage organoids provide a unique platform to study developmental cues guiding endothelial and mesenchymal cell fate determination, and investigate intricate cell-cell communications in human organogenesis and disease. Highlights: BMP signaling fine-tunes the co-differentiation of mesoderm and endoderm.The cellular composition in multilineage organoids resembles that of human fetal organs.Mesenchyme and endothelium co-developed within the organoids adopt organ-specific characteristics.Multilineage organoids recapitulate abnormal endothelial-epithelial crosstalk in FOXF1-associated disorders.

2.
Am J Respir Crit Care Med ; 208(6): 709-725, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37463497

RESUMO

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal developmental disorder of lung morphogenesis caused by insufficiency of FOXF1 (forkhead box F1) transcription factor function. The cellular and transcriptional mechanisms by which FOXF1 deficiency disrupts human lung formation are unknown. Objectives: To identify cell types, gene networks, and cell-cell interactions underlying the pathogenesis of ACDMPV. Methods: We used single-nucleus RNA and assay for transposase-accessible chromatin sequencing, immunofluorescence confocal microscopy, and RNA in situ hybridization to identify cell types and molecular networks influenced by FOXF1 in ACDMPV lungs. Measurements and Main Results: Pathogenic single-nucleotide variants and copy-number variant deletions involving the FOXF1 gene locus in all subjects with ACDMPV (n = 6) were accompanied by marked changes in lung structure, including deficient alveolar development and a paucity of pulmonary microvasculature. Single-nucleus RNA and assay for transposase-accessible chromatin sequencing identified alterations in cell number and gene expression in endothelial cells (ECs), pericytes, fibroblasts, and epithelial cells in ACDMPV lungs. Distinct cell-autonomous roles for FOXF1 in capillary ECs and pericytes were identified. Pathogenic variants involving the FOXF1 gene locus disrupt gene expression in EC progenitors, inhibiting the differentiation or survival of capillary 2 ECs and cell-cell interactions necessary for both pulmonary vasculogenesis and alveolar type 1 cell differentiation. Loss of the pulmonary microvasculature was associated with increased VEGFA (vascular endothelial growth factor A) signaling and marked expansion of systemic bronchial ECs expressing COL15A1 (collagen type XV α 1 chain). Conclusions: Distinct FOXF1 gene regulatory networks were identified in subsets of pulmonary endothelial and fibroblast progenitors, providing both cellular and molecular targets for the development of therapies for ACDMPV and other diffuse lung diseases of infancy.


Assuntos
Síndrome da Persistência do Padrão de Circulação Fetal , Recém-Nascido , Humanos , Síndrome da Persistência do Padrão de Circulação Fetal/genética , Síndrome da Persistência do Padrão de Circulação Fetal/patologia , Redes Reguladoras de Genes/genética , Fator A de Crescimento do Endotélio Vascular/genética , Células Endoteliais/patologia , Multiômica , Pulmão/patologia , RNA , Fatores de Transcrição Forkhead/genética
3.
Semin Perinatol ; 47(3): 151729, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37012138

RESUMO

Neonatal lung and heart diseases, albeit rare, can result in poor quality of life, often require long-term management and/or organ transplantation. For example, Congenital Heart Disease (CHD) is one of the most common type of congenital disabilities, affecting nearly 1% of the newborns, and has complex and multifactorial causes, including genetic predisposition and environmental influences. To develop new strategies for heart and lung regeneration in CHD and neonatal lung disease, human induced pluripotent stem cells (hiPSCs) provide a unique and personalized platform for future cell replacement therapy and high-throughput drug screening. Additionally, given the differentiation potential of iPSCs, cardiac cell types such as cardiomyocytes, endothelial cells, and fibroblasts and lung cell types such Type II alveolar epithelial cells can be derived in a dish to study the fundamental pathology during disease progression. In this review, we discuss the applications of hiPSCs in understanding the molecular mechanisms and cellular phenotypes of CHD (e.g., structural heart defect, congenital valve disease, and congenital channelopathies) and congenital lung diseases, such as surfactant deficiencies and Brain-Lung-Thyroid syndrome. We also provide future directions for generating mature cell types from iPSCs, and more complex hiPSC-based systems using three-dimensional (3D) organoids and tissue-engineering. With these potential advancements, the promise that hiPSCs will deliver new CHD and neonatal lung disease treatments may soon be fulfilled.


Assuntos
Cardiopatias Congênitas , Células-Tronco Pluripotentes Induzidas , Doenças do Recém-Nascido , Pneumopatias , Recém-Nascido , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Endoteliais , Qualidade de Vida , Cardiopatias Congênitas/terapia , Desenvolvimento de Medicamentos , Pneumopatias/terapia , Pneumopatias/metabolismo
4.
J Nanobiotechnology ; 21(1): 40, 2023 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-36739414

RESUMO

Cancer immunotherapy shows promising potential for treating breast cancer. While patients may have heterogeneous treatment responses for adjuvant therapy, it is challenging to predict an individual patient's response to cancer immunotherapy. Here, we report primary tumor-derived organotypic cell clusters (POCCs) for rapid and reliable evaluation of cancer immunotherapy. By using a label-free, contactless, and highly biocompatible acoustofluidic method, hundreds of cell clusters could be assembled from patient primary breast tumor dissociation within 2 min. Through the incorporation of time-lapse living cell imaging, the POCCs could faithfully recapitulate the cancer-immune interaction dynamics as well as their response to checkpoint inhibitors. Superior to current tumor organoids that usually take more than two weeks to develop, the POCCs can be established and used for evaluation of cancer immunotherapy within 12 h. The POCCs can preserve the cell components from the primary tumor due to the short culture time. Moreover, the POCCs can be assembled with uniform fabricate size and cell composition and served as an open platform for manipulating cell composition and ratio under controlled treatment conditions with a short turnaround time. Thus, we provide a new method to identify potentially immunogenic breast tumors and test immunotherapy, promoting personalized cancer therapy.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/terapia , Imunoterapia/métodos
5.
Am J Respir Crit Care Med ; 207(8): 1042-1054, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-36480964

RESUMO

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is linked to heterozygous mutations in the FOXF1 (Forkhead Box F1) gene, a key transcriptional regulator of pulmonary vascular development. There are no effective treatments for ACDMPV other than lung transplant, and new pharmacological agents activating FOXF1 signaling are urgently needed. Objectives: Identify-small molecule compounds that stimulate FOXF1 signaling. Methods: We used mass spectrometry, immunoprecipitation, and the in vitro ubiquitination assay to identify TanFe (transcellular activator of nuclear FOXF1 expression), a small-molecule compound from the nitrile group, which stabilizes the FOXF1 protein in the cell. The efficacy of TanFe was tested in mouse models of ACDMPV and acute lung injury and in human vascular organoids derived from induced pluripotent stem cells of a patient with ACDMPV. Measurements and Main Results: We identified HECTD1 as an E3 ubiquitin ligase involved in ubiquitination and degradation of the FOXF1 protein. The TanFe compound disrupted FOXF1-HECTD1 protein-protein interactions and decreased ubiquitination of the FOXF1 protein in pulmonary endothelial cells in vitro. TanFe increased protein concentrations of FOXF1 and its target genes Flk1, Flt1, and Cdh5 in LPS-injured mouse lungs, decreasing endothelial permeability and inhibiting lung inflammation. Treatment of pregnant mice with TanFe increased FOXF1 protein concentrations in lungs of Foxf1+/- embryos, stimulated neonatal lung angiogenesis, and completely prevented the mortality of Foxf1+/- mice after birth. TanFe increased angiogenesis in human vascular organoids derived from induced pluripotent stem cells of a patient with ACDMPV with FOXF1 deletion. Conclusions: TanFe is a novel activator of FOXF1, providing a new therapeutic candidate for treatment of ACDMPV and other neonatal pulmonary vascular diseases.


Assuntos
Síndrome da Persistência do Padrão de Circulação Fetal , Recém-Nascido , Humanos , Animais , Camundongos , Síndrome da Persistência do Padrão de Circulação Fetal/genética , Células Endoteliais , Pulmão/metabolismo , Fatores de Transcrição Forkhead/genética
6.
Adv Sci (Weinh) ; 9(27): e2200475, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35908805

RESUMO

The aging of the immune system drives systemic aging and the pathogenesis of age-related diseases. However, a significant knowledge gap remains in understanding immune-driven aging, especially in brain aging, due to the limited current in vitro models of neuroimmune interaction. Here, the authors report the development of a human brain organoid microphysiological analysis platform (MAP) to discover the dynamic process of immune-driven brain aging. The organoid MAP is created by 3D printing that confines organoid growth and facilitates cell and nutrition perfusion, promoting organoid maturation and their committment to forebrain identity. Dynamic rocking flow is incorporated into the platform that allows to perfuse primary monocytes from young (20 to 30-year-old) and aged (>60-year-old) donors and culture human cortical organoids to model neuroimmune interaction. The authors find that the aged monocytes increase infiltration and promote the expression of aging-related markers (e.g., higher expression of p16) within the human cortical organoids, indicating that aged monocytes may drive brain aging. The authors believe that the organoid MAP may provide promising solutions for basic research and translational applications in aging, neural immunological diseases, autoimmune disorders, and cancer.


Assuntos
Neoplasias , Organoides , Adulto , Envelhecimento , Encéfalo , Humanos , Imunoterapia , Pessoa de Meia-Idade , Adulto Jovem
7.
Theranostics ; 12(8): 3628-3636, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35664082

RESUMO

Rationale: Predicting tumor responses to adjuvant therapies can potentially help guide treatment decisions and improve patient survival. Currently, tumor pathology, histology, and molecular profiles are being integrated into personalized profiles to guide therapeutic decisions. However, it remains a grand challenge to evaluate tumor responses to immunotherapy for personalized medicine. Methods: We present a microfluidics-based mini-tumor chip approach to predict tumor responses to cancer immunotherapy in a preclinical model. By uniformly infusing dissociated tumor cells into isolated microfluidic well-arrays, 960 mini-tumors could be uniformly generated on-chip, with each well representing the ex vivo tumor niche that preserves the original tumor cell composition and dynamic cell-cell interactions and autocrine/paracrine cytokines. Results: By incorporating time-lapse live-cell imaging, our mini-tumor chip allows the investigation of dynamic immune-tumor interactions as well as their responses to cancer immunotherapy (e.g., anti-PD1 treatment) in parallel within 36 hours. Additionally, by establishing orthotopic breast tumor models with constitutive differential PD-L1 expression levels, we showed that the on-chip interrogation of the primary tumor's responses to anti-PD1 as early as 10 days post tumor inoculation could predict the in vivo tumors' responses to anti-PD1 at the endpoint of day 24. We also demonstrated the application of this mini-tumor chip to interrogate on-chip responses of primary tumor cells isolated from primary human breast and renal tumor tissues. Conclusions: Our approach provides a simple, quick-turnaround solution to measure tumor responses to cancer immunotherapy.


Assuntos
Imunoterapia , Neoplasias , Terapia Combinada , Humanos , Imunoterapia/métodos , Microfluídica , Neoplasias/terapia , Medicina de Precisão/métodos , Microambiente Tumoral
8.
Adv Sci (Weinh) ; 9(22): e2201478, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35611994

RESUMO

Tumor microenvironment crosstalk, in particular interactions between cancer cells, T cells, and myeloid-derived suppressor cells (MDSCs), mediates tumor initiation, progression, and response to treatment. However, current patient-derived models such as tumor organoids and 2D cultures lack some essential niche cell types (e.g., MDSCs) and fail to model complex tumor-immune interactions. Here, the authors present the novel acoustically assembled patient-derived cell clusters (APCCs) that can preserve original tumor/immune cell compositions, model their interactions in 3D microenvironments, and test the treatment responses of primary tumors in a rapid, scalable, and user-friendly manner. By incorporating a large array of 3D acoustic trappings within the extracellular matrix, hundreds of APCCs can be assembled within a petri dish within 2 min. Moreover, the APCCs can preserve sensitive and short-lived (≈1 to 2-day lifespan in vivo) tumor-induced MDSCs and model their dynamic suppression of T cell tumor toxicity for up to 24 h. Finally, using the APCCs, the authors succesully model the combinational therapeutic effect of a multi-kinase inhibitor targeting MDSCs (cabozantinib) and an anti-PD-1 immune checkpoint inhibitor (pembrolizumab). The novel APCCs may hold promising potential in predicting treatment response for personalized cancer adjuvant therapy as well as screening novel cancer immunotherapy and combinational therapy.


Assuntos
Células Supressoras Mieloides , Neoplasias , Humanos , Inibidores de Checkpoint Imunológico , Imunoterapia , Células Supressoras Mieloides/metabolismo , Neoplasias/terapia , Microambiente Tumoral
9.
Sci Transl Med ; 14(638): eabl8574, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35353543

RESUMO

Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque (Macaca mulatta) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1ß and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.


Assuntos
Corioamnionite , Nascimento Prematuro , Animais , Corioamnionite/induzido quimicamente , Corioamnionite/patologia , Feminino , Pulmão/patologia , Macaca mulatta , Gravidez , Nascimento Prematuro/prevenção & controle , Troca Gasosa Pulmonar
10.
Expert Opin Ther Targets ; 25(8): 621-632, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34488532

RESUMO

INTRODUCTION: Hypoplastic left heart syndrome (HLHS) is a severe developmental defect characterized by the underdevelopment of the left ventricle along with aortic and valvular defects. Multiple palliative surgeries are required for survival. Emerging studies have identified potential mechanisms for the disease onset, including genetic and hemodynamic causes. Genetic variants associated with HLHS include transcription factors, chromatin remodelers, structural proteins, and signaling proteins necessary for normal heart development. Nonetheless, current therapies are being tested clinically and have shown promising results at improving cardiac function in patients who have undergone palliative surgeries. AREAS COVERED: We searched PubMed and clinicaltrials.gov to review most of the mechanistic research and clinical trials involving HLHS. This review discusses the anatomy and pathology of HLHS hearts. We highlight some of the identified genetic variants that underly the molecular pathogenesis of HLHS. Additionally, we discuss some of the emerging therapies and their limitations for HLHS. EXPERT OPINION: While HLHS etiology is largely obscure, palliative therapies remain the most viable option for the patients. It is necessary to generate animal and stem cell models to understand the underlying genetic causes directly leading to HLHS and facilitate the use of gene-based therapies to improve cardiac development and regeneration.


Assuntos
Síndrome do Coração Esquerdo Hipoplásico , Animais , Humanos , Síndrome do Coração Esquerdo Hipoplásico/tratamento farmacológico , Síndrome do Coração Esquerdo Hipoplásico/genética , Regeneração/genética , Fatores de Transcrição
11.
Nat Commun ; 11(1): 1673, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32245974

RESUMO

Environmental and epigenetic factors often play an important role in polygenic disorders. However, how such factors affect disease-specific tissues at the molecular level remains to be understood. Here, we address this in pulmonary arterial hypertension (PAH). We obtain pulmonary arterial endothelial cells (PAECs) from lungs of patients and controls (n = 19), and perform chromatin, transcriptomic and interaction profiling. Overall, we observe extensive remodeling at active enhancers in PAH PAECs and identify hundreds of differentially active TFs, yet find very little transcriptomic changes in steady-state. We devise a disease-specific enhancer-gene regulatory network and predict that primed enhancers in PAH PAECs are activated by the differentially active TFs, resulting in an aberrant response to endothelial signals, which could lead to disturbed angiogenesis and endothelial-to-mesenchymal-transition. We validate these predictions for a selection of target genes in PAECs stimulated with TGF-ß, VEGF or serotonin. Our study highlights the role of chromatin state and enhancers in disease-relevant cell types of PAH.


Assuntos
Elementos Facilitadores Genéticos , Redes Reguladoras de Genes , Hipertensão Arterial Pulmonar/genética , Artéria Pulmonar/patologia , Remodelação Vascular/genética , Adulto , Biópsia , Estudos de Casos e Controles , Células Cultivadas , Cromatina/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/citologia , Epigênese Genética , Transição Epitelial-Mesenquimal/genética , Feminino , Código das Histonas/genética , Histonas/genética , Humanos , Lactente , Pulmão/irrigação sanguínea , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , Hipertensão Arterial Pulmonar/patologia , Artéria Pulmonar/citologia , RNA-Seq , Serotonina/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
12.
Exp Mol Med ; 50(4): 1-7, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29651007

RESUMO

Life-threatening malignant arrhythmias in pathophysiological conditions can increase the mortality and morbidity of patients with cardiovascular diseases. Cardiac electrical activity depends on the coordinated propagation of excitatory stimuli and the generation of action potentials in cardiomyocytes. Action potential formation results from the opening and closing of ion channels. Recent studies have indicated that small-conductance calcium-activated potassium (SK) channels play a critical role in cardiac repolarization in pathophysiological but not normal physiological conditions. The aim of this review is to describe the role of SK channels in healthy and diseased hearts, to suggest cardiovascular pathophysiologic targets for intervention, and to discuss studies of agents that target SK channels for the treatment of cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Miocárdio/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Animais , Doenças Cardiovasculares/fisiopatologia , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Humanos , Mutação , Canais de Potássio Cálcio-Ativados/química , Canais de Potássio Cálcio-Ativados/genética , Relação Estrutura-Atividade
13.
Circulation ; 136(20): 1920-1935, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-28935667

RESUMO

BACKGROUND: Immune dysregulation has been linked to occlusive vascular remodeling in pulmonary arterial hypertension (PAH) that is hereditary, idiopathic, or associated with other conditions. Circulating autoantibodies, lung perivascular lymphoid tissue, and elevated cytokines have been related to PAH pathogenesis but without a clear understanding of how these abnormalities are initiated, perpetuated, and connected in the progression of disease. We therefore set out to identify specific target antigens in PAH lung immune complexes as a starting point toward resolving these issues to better inform future application of immunomodulatory therapies. METHODS: Lung immune complexes were isolated and PAH target antigens were identified by liquid chromatography tandem mass spectrometry, confirmed by enzyme-linked immunosorbent assay, and localized by confocal microscopy. One PAH antigen linked to immunity and inflammation was pursued and a link to PAH pathophysiology was investigated by next-generation sequencing, functional studies in cultured monocytes and endothelial cells, and hemodynamic and lung studies in a rat. RESULTS: SAM domain and HD domain-containing protein 1 (SAMHD1), an innate immune factor that suppresses HIV replication, was identified and confirmed as highly expressed in immune complexes from 16 hereditary and idiopathic PAH versus 12 control lungs. Elevated SAMHD1 was localized to endothelial cells, perivascular dendritic cells, and macrophages, and SAMHD1 antibodies were prevalent in tertiary lymphoid tissue. An unbiased screen using metagenomic sequencing related SAMHD1 to increased expression of human endogenous retrovirus K (HERV-K) in PAH versus control lungs (n=4). HERV-K envelope and deoxyuridine triphosphate nucleotidohydrolase mRNAs were elevated in PAH versus control lungs (n=10), and proteins were localized to macrophages. HERV-K deoxyuridine triphosphate nucleotidohydrolase induced SAMHD1 and proinflammatory cytokines (eg, interleukin 6, interleukin 1ß, and tumor necrosis factor α) in circulating monocytes, pulmonary arterial endothelial cells, and also activated B cells. Vulnerability of pulmonary arterial endothelial cells (PAEC) to apoptosis was increased by HERV-K deoxyuridine triphosphate nucleotidohydrolase in an interleukin 6-independent manner. Furthermore, 3 weekly injections of HERV-K deoxyuridine triphosphate nucleotidohydrolase induced hemodynamic and vascular changes of pulmonary hypertension in rats (n=8) and elevated interleukin 6. CONCLUSIONS: Our study reveals that upregulation of the endogenous retrovirus HERV-K could both initiate and sustain activation of the immune system and cause vascular changes associated with PAH.


Assuntos
Hipertensão Pulmonar/imunologia , Mediadores da Inflamação/imunologia , Regulação para Cima/fisiologia , Proteínas Virais/biossíntese , Proteínas Virais/imunologia , Adolescente , Adulto , Animais , Complexo Antígeno-Anticorpo/biossíntese , Complexo Antígeno-Anticorpo/imunologia , Células Cultivadas , Criança , Técnicas de Cocultura , Feminino , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Lactente , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Proteína 1 com Domínio SAM e Domínio HD/biossíntese , Proteína 1 com Domínio SAM e Domínio HD/imunologia , Adulto Jovem
14.
Circulation ; 133(18): 1783-94, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27045138

RESUMO

BACKGROUND: We previously reported high-throughput RNA sequencing analyses that identified heightened expression of the chromatin architectural factor High Mobility Group AT-hook 1 (HMGA1) in pulmonary arterial endothelial cells (PAECs) from patients who had idiopathic pulmonary arterial hypertension (PAH) in comparison with controls. Because HMGA1 promotes epithelial-to-mesenchymal transition in cancer, we hypothesized that increased HMGA1 could induce transition of PAECs to a smooth muscle (SM)-like mesenchymal phenotype (endothelial-to-mesenchymal transition), explaining both dysregulation of PAEC function and possible cellular contribution to the occlusive remodeling that characterizes advanced idiopathic PAH. METHODS AND RESULTS: We documented increased HMGA1 in PAECs cultured from idiopathic PAH versus donor control lungs. Confocal microscopy of lung explants localized the increase in HMGA1 consistently to pulmonary arterial endothelium, and identified many cells double-positive for HMGA1 and SM22α in occlusive and plexogenic lesions. Because decreased expression and function of bone morphogenetic protein receptor 2 (BMPR2) is observed in PAH, we reduced BMPR2 by small interfering RNA in control PAECs and documented an increase in HMGA1 protein. Consistent with transition of PAECs by HMGA1, we detected reduced platelet endothelial cell adhesion molecule 1 (CD31) and increased endothelial-to-mesenchymal transition markers, αSM actin, SM22α, calponin, phospho-vimentin, and Slug. The transition was associated with spindle SM-like morphology, and the increase in αSM actin was largely reversed by joint knockdown of BMPR2 and HMGA1 or Slug. Pulmonary endothelial cells from mice with endothelial cell-specific loss of Bmpr2 showed similar gene and protein changes. CONCLUSIONS: Increased HMGA1 in PAECs resulting from dysfunctional BMPR2 signaling can transition endothelium to SM-like cells associated with PAH.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/deficiência , Transição Epitelial-Mesenquimal/fisiologia , Proteína HMGA1a/biossíntese , Hipertensão Pulmonar/metabolismo , Fatores de Transcrição da Família Snail/biossíntese , Adolescente , Adulto , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Células Cultivadas , Criança , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Proteína HMGA1a/genética , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Lactente , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fatores de Transcrição da Família Snail/genética , Adulto Jovem
15.
Eur Heart J ; 36(13): 806-16, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25368203

RESUMO

AIMS: High-fat diet-induced obesity (DIO) is a major contributor to type II diabetes and micro- and macro-vascular complications leading to peripheral vascular disease (PVD). Metabolic abnormalities of induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) from obese individuals could potentially limit their therapeutic efficacy for PVD. The aim of this study was to compare the function of iPSC-ECs from normal and DIO mice using comprehensive in vitro and in vivo assays. METHODS AND RESULTS: Six-week-old C57Bl/6 mice were fed with a normal or high-fat diet. At 24 weeks, iPSCs were generated from tail tip fibroblasts and differentiated into iPSC-ECs using a directed monolayer approach. In vitro functional analysis revealed that iPSC-ECs from DIO mice had significantly decreased capacity to form capillary-like networks, diminished migration, and lower proliferation. Microarray and ELISA confirmed elevated apoptotic, inflammatory, and oxidative stress pathways in DIO iPSC-ECs. Following hindlimb ischaemia, mice receiving intramuscular injections of DIO iPSC-ECs had significantly decreased reperfusion compared with mice injected with control healthy iPSC-ECs. Hindlimb sections revealed increased muscle atrophy and presence of inflammatory cells in mice receiving DIO iPSC-ECs. When pravastatin was co-administered to mice receiving DIO iPSC-ECs, a significant increase in reperfusion was observed; however, this beneficial effect was blunted by co-administration of the nitric oxide synthase inhibitor, N(ω)-nitro-l-arginine methyl ester. CONCLUSION: This is the first study to provide evidence that iPSC-ECs from DIO mice exhibit signs of endothelial dysfunction and have suboptimal efficacy following transplantation in a hindlimb ischaemia model. These findings may have important implications for future treatment of PVD using iPSC-ECs in the obese population.


Assuntos
Células Endoteliais/fisiologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Óxido Nítrico/fisiologia , Obesidade/fisiopatologia , Pravastatina/farmacologia , Análise de Variância , Animais , Apoptose/fisiologia , Diferenciação Celular , Dieta Hiperlipídica , Inibidores Enzimáticos/farmacologia , Fibroblastos/fisiologia , Membro Posterior/irrigação sanguínea , Injeções Intramusculares , Isquemia/fisiopatologia , Isquemia/prevenção & controle , Camundongos Endogâmicos C57BL , Músculo Esquelético , Doenças Musculares/prevenção & controle , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Distribuição Aleatória , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais
16.
Circ Res ; 111(7): 882-93, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22821929

RESUMO

RATIONALE: Induced pluripotent stem cells (iPSCs) hold great promise for the development of patient-specific therapies for cardiovascular disease. However, clinical translation will require preclinical optimization and validation of large-animal iPSC models. OBJECTIVE: To successfully derive endothelial cells from porcine iPSCs and demonstrate their potential utility for the treatment of myocardial ischemia. METHODS AND RESULTS: Porcine adipose stromal cells were reprogrammed to generate porcine iPSCs (piPSCs). Immunohistochemistry, quantitative PCR, microarray hybridization, and angiogenic assays confirmed that piPSC-derived endothelial cells (piPSC-ECs) shared similar morphological and functional properties as endothelial cells isolated from the autologous pig aorta. To demonstrate their therapeutic potential, piPSC-ECs were transplanted into mice with myocardial infarction. Compared with control, animals transplanted with piPSC-ECs showed significant functional improvement measured by echocardiography (fractional shortening at week 4: 27.2±1.3% versus 22.3±1.1%; P<0.001) and MRI (ejection fraction at week 4: 45.8±1.3% versus 42.3±0.9%; P<0.05). Quantitative protein assays and microfluidic single-cell PCR profiling showed that piPSC-ECs released proangiogenic and antiapoptotic factors in the ischemic microenvironment, which promoted neovascularization and cardiomyocyte survival, respectively. Release of paracrine factors varied significantly among subpopulations of transplanted cells, suggesting that transplantation of specific cell populations may result in greater functional recovery. CONCLUSIONS: In summary, this is the first study to successfully differentiate piPSCs-ECs from piPSCs and demonstrate that transplantation of piPSC-ECs improved cardiac function after myocardial infarction via paracrine activation. Further development of these large animal iPSC models will yield significant insights into their therapeutic potential and accelerate the clinical translation of autologous iPSC-based therapy.


Assuntos
Transplante de Células , Endotélio Vascular/citologia , Endotélio Vascular/transplante , Coração/fisiopatologia , Técnicas Analíticas Microfluídicas , Infarto do Miocárdio/terapia , Comunicação Parácrina/fisiologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Ecocardiografia , Endotélio Vascular/fisiologia , Feminino , Imageamento por Ressonância Magnética , Camundongos , Camundongos SCID , Modelos Animais , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Neovascularização Fisiológica , Células-Tronco Pluripotentes/fisiologia , Suínos , Porco Miniatura
17.
J Proteome Res ; 11(4): 2365-73, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22428589

RESUMO

As inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase, statins have pleiotropic vascular-protective effects, such as anti-inflammatory and antioxidative effects. We investigated the short-term beneficial effects of statins on modulating the translocation of lipid-raft-related proteins in endothelial cells (ECs). Human umbilical vein ECs were treated with atorvastatin for 30 min or 2 h; lipid-raft proteins were isolated and examined by quantitative proteome assay. Functional classification of identified proteins in lipid rafts revealed upregulated antioxidative proteins; downregulated proteins were associated with inflammation and cell adhesion. Among proteins verified by Western blot analysis, endoplasmic reticulum protein 46 (ERp46) showed increased level in lipid rafts with atorvastatin. Further, atorvastatin inhibited the activation of membrane-bound NADPH oxidase in both untreated and angiotensin II-treated ECs, as shown by reduced reactive oxygen species production. Co-immunoprecipitation and immunofluorescence experiments revealed that atorvastatin increased the association of ERp46 and Nox2, an NADPH oxidase isoform, in lipid rafts, thereby inhibiting Nox2 assembly with its regulatory subunits, such as p47phox and p67phox. Our results reveal a novel antioxidative role of atorvastatin by promoting the membrane translocation of ERp46 and its binding with Nox2 to inhibit Nox2 activity in ECs, which may offer another insight into the pleiotropic functions of statins.


Assuntos
Antioxidantes/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Microdomínios da Membrana/química , Proteoma/metabolismo , Sequência de Aminoácidos , Atorvastatina , Ácidos Heptanoicos/farmacologia , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/metabolismo , Dados de Sequência Molecular , NADPH Oxidase 2 , NADPH Oxidases/análise , NADPH Oxidases/metabolismo , Isomerases de Dissulfetos de Proteínas/análise , Isomerases de Dissulfetos de Proteínas/metabolismo , Proteínas/análise , Proteínas/classificação , Proteínas/metabolismo , Proteoma/análise , Proteoma/efeitos dos fármacos , Pirróis/farmacologia , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo , Reprodutibilidade dos Testes
18.
Circ Res ; 108(4): 410-7, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21193741

RESUMO

RATIONALE: Endothelial cells (ECs) have distinct mechanotransduction mechanisms responding to laminar versus disturbed flow patterns. Endothelial dysfunction, affected by imposed flow, is one of the earliest events leading to atherogenesis. The involvement of γ/δ T lymphocytes in endothelial dysfunction under flow is largely unknown. OBJECTIVE: To investigate whether shear stress regulates membrane translocation of ATP synthase ß chain (ATPSß) in ECs, leading to the increased γ/δ T-lymphocyte adhesion and the related functions. METHOD AND RESULTS: We applied different flow patterns to cultured ECs. Laminar flow decreased the level of membrane-bound ATPSß (ecto-ATPSß) and depleted membrane cholesterol, whereas oscillatory flow increased the level of ecto-ATPSß and membrane cholesterol. Incubating ECs with cholesterol or depleting cellular cholesterol with ß-cyclodextrin mimicked the effect of oscillatory or laminar flow, respectively. Knockdown caveolin-1 by small interfering RNA prevented ATPSß translocation in response to laminar flow. Importantly, oscillatory flow or cholesterol treatment elevated the number of γ/δ T cells binding to ECs, which was blocked by anti-ATPSß antibody. Furthermore, the incubation of γ/δ T cells with ECs increased tumor necrosis fact α and interferon-γ secretion from T cells and vascular cell adhesion molecule-1 expression in ECs. In vivo, γ/δ T-cell adhesion and ATPSß membrane translocation was elevated in the aortic inner curvature and disturbed flow areas in partially ligated carotid arteries of ApoE(-/-) mice fed a high-fat diet. CONCLUSIONS: This study provides evidence that disturbed flow and hypercholesterolemia synergistically promote γ/δ T-lymphocyte activation by the membrane translocation of ATPSß in ECs and in vivo in mice, which is a novel mechanism of endothelial activation.


Assuntos
Comunicação Celular/fisiologia , Endotélio Vascular/citologia , ATPases Mitocondriais Próton-Translocadoras/fisiologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Estresse Mecânico , Linfócitos T/citologia , Linfócitos T/metabolismo , Adenosina Trifosfatases/fisiologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Transporte Biológico/fisiologia , Caveolina 1/fisiologia , Adesão Celular/fisiologia , Membrana Celular/fisiologia , Células Cultivadas , Colesterol/metabolismo , Endotélio Vascular/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA