Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Small ; : e2305940, 2023 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-37803920

RESUMO

Nanomedicine technologies are being developed for the prevention, diagnosis, and treatment of cardiovascular disease (CVD), which is the leading cause of death worldwide. Before delving into the nuances of cardiac nanomedicine, it is essential to comprehend the fundamental sex-specific differences in cardiovascular health. Traditionally, CVDs have been more prevalent in males, but it is increasingly evident that females also face significant risks, albeit with distinct characteristics. Females tend to develop CVDs at a later age, exhibit different clinical symptoms, and often experience worse outcomes compared to males. These differences indicate the need for sex-specific approaches in cardiac nanomedicine. This Perspective discusses the importance of considering sex in the safety and therapeutic efficacy of nanomedicine approaches for the prevention, diagnosis, and treatment of CVD.

3.
Opt Lett ; 48(15): 3929-3932, 2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-37527085

RESUMO

To reveal the three-dimensional microstructure and calcium dynamics of human heart organoids (hHOs), we developed a dual-modality imaging system combining the advantages of optical coherence tomography (OCT) and fluorescence microscopy. OCT provides high-resolution volumetric structural information, while fluorescence imaging indicates the electrophysiology of the hHOs' beating behavior. We verified that concurrent OCT motion mode (M-mode) and calcium imaging retrieved the same beating pattern from the heart organoids. We further applied dynamic contrast OCT (DyC-OCT) analysis to strengthen the verification and localize the beating clusters inside the hHOs. This imaging platform provides a powerful tool for studying and assessing hHOs in vitro, with potential applications in disease modeling and drug screening.


Assuntos
Cálcio , Coração , Humanos , Coração/diagnóstico por imagem , Microscopia de Fluorescência , Tomografia de Coerência Óptica/métodos , Organoides/diagnóstico por imagem
4.
EMBO Rep ; 20(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30886000

RESUMO

Cardiac dysfunctions dramatically increase with age. Revealing a currently unknown contributor to cardiac ageing, we report the age-dependent, cardiac-specific accumulation of the lysosphingolipid sphinganine (dihydrosphingosine, DHS) as an evolutionarily conserved hallmark of the aged vertebrate heart. Mechanistically, the DHS-derivative sphinganine-1-phosphate (DHS1P) directly inhibits HDAC1, causing an aberrant elevation in histone acetylation and transcription levels, leading to DNA damage. Accordingly, the pharmacological interventions, preventing (i) the accumulation of DHS1P using SPHK2 inhibitors, (ii) the aberrant increase in histone acetylation using histone acetyltransferase (HAT) inhibitors, (iii) the DHS1P-dependent increase in transcription using an RNA polymerase II inhibitor, block DHS-induced DNA damage in human cardiomyocytes. Importantly, an increase in DHS levels in the hearts of healthy young adult mice leads to an impairment in cardiac functionality indicated by a significant reduction in left ventricular fractional shortening and ejection fraction, mimicking the functional deterioration of aged hearts. These molecular and functional defects can be partially prevented in vivo using HAT inhibitors. Together, we report an evolutionarily conserved mechanism by which increased DHS levels drive the decline in cardiac health.


Assuntos
Envelhecimento/genética , Envelhecimento/metabolismo , Variação Genética , Instabilidade Genômica , Miocárdio/metabolismo , Esfingolipídeos/metabolismo , Animais , Curcumina/química , Curcumina/farmacologia , Dano ao DNA/efeitos dos fármacos , Metabolismo Energético , Epigênese Genética , Evolução Molecular , Fundulidae , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genômica/métodos , Histona Acetiltransferases/química , Histona Acetiltransferases/metabolismo , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Humanos , Modelos Moleculares , Miócitos Cardíacos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Relação Estrutura-Atividade , Vertebrados/genética , Vertebrados/metabolismo
6.
Clin Exp Hypertens ; 40(2): 141-149, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28783384

RESUMO

The main purpose of this study was to determine some key physical, physiological, clinical, and nutritional markers of health status in obese and sedentary adults (54.0 ± 8.1 years, 141 men and 68 women) with primary hypertension (HTN) characterized by sex and cardiorespiratory fitness (CRF) level. The studied population showed a high cardiovascular risk (CVR) profile including metabolically abnormal obese, with poor CRF level (22.5 ± 5.6 mL·kg-1·min-1), exercise-induced HTN (Systolic Blood Pressure>210 mmHg in men and >190 mmHg in women at the end of the exercise test) and with non-healthy adherence to dietary pattern (Dietary Approaches to Stop Hypertension, 46.3%; Mediterranean Diet, 41.1%; and Healthy Diet Indicator, 37.1%). Women showed a better biochemical and dietary pattern profile than men (lower values, P < 0.05, in triglycerides, mean difference = 26.3; 95% CI = 0.9-51.7 mg/dL, aspartate transaminase, mean difference = 4.2; 95% CI = 0.3-8.0 U/L; alanine transaminase, mean difference = 8.2; 95% CI = 1.6-14.8 U/L; gamma-glutamyl transpeptidase, mean difference = 11.0; 95% CI = -1.1-23.2 U/L and higher values, P = 0.002, in high-density lipoprotein cholesterol, mean difference = 5.0, 95% CI = -13.3-3.3 mg/dL), but physical and peak exercise physiological characteristics were poorer. A higher CRF level might contribute to the attenuation of some CVR factors, such as high body mass index, non-dipping profile, and high hepatic fat. The results strongly suggest that targeting key behaviors such as improving nutritional quality and CRF via regular physical activity will contribute to improving the health with independent beneficial effects on CVR factors.


Assuntos
Aptidão Cardiorrespiratória , Dieta Saudável , Hipertensão/fisiopatologia , Obesidade/fisiopatologia , Cooperação do Paciente , Adulto , Idoso , Alanina Transaminase/sangue , Aspartato Aminotransferases/sangue , Pressão Sanguínea , Índice de Massa Corporal , HDL-Colesterol/sangue , Teste de Esforço , Feminino , Nível de Saúde , Humanos , Hipertensão/complicações , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Esforço Físico/fisiologia , Fatores de Risco , Comportamento Sedentário , Fatores Sexuais , Triglicerídeos/sangue , Adulto Jovem , gama-Glutamiltransferase/sangue
7.
Circulation ; 131(14): 1278-1290, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25739401

RESUMO

BACKGROUND: Long noncoding RNAs (lncRNAs) have emerged as critical epigenetic regulators with important functions in development and disease. Here, we sought to identify and functionally characterize novel lncRNAs critical for vertebrate development. METHODS AND RESULTS: By relying on human pluripotent stem cell differentiation models, we investigated lncRNAs differentially regulated at key steps during human cardiovascular development with a special focus on vascular endothelial cells. RNA sequencing led to the generation of large data sets that serve as a gene expression roadmap highlighting gene expression changes during human pluripotent cell differentiation. Stage-specific analyses led to the identification of 3 previously uncharacterized lncRNAs, TERMINATOR, ALIEN, and PUNISHER, specifically expressed in undifferentiated pluripotent stem cells, cardiovascular progenitors, and differentiated endothelial cells, respectively. Functional characterization, including localization studies, dynamic expression analyses, epigenetic modification monitoring, and knockdown experiments in lower vertebrates, as well as murine embryos and human cells, confirmed a critical role for each lncRNA specific for each analyzed developmental stage. CONCLUSIONS: We have identified and functionally characterized 3 novel lncRNAs involved in vertebrate and human cardiovascular development, and we provide a comprehensive transcriptomic roadmap that sheds new light on the molecular mechanisms underlying human embryonic development, mesodermal commitment, and cardiovascular specification.


Assuntos
Sistema Cardiovascular/crescimento & desenvolvimento , Células Endoteliais/química , Regulação da Expressão Gênica no Desenvolvimento/genética , Miócitos Cardíacos/química , Células-Tronco Pluripotentes/química , RNA Longo não Codificante/isolamento & purificação , Vertebrados/genética , Animais , Sistema Cardiovascular/metabolismo , Diferenciação Celular , Linhagem da Célula , Mapeamento Cromossômico , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Coração Fetal/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Dados de Sequência Molecular , Morfolinos/farmacocinética , Miócitos Cardíacos/citologia , RNA Longo não Codificante/fisiologia , Análise de Sequência de RNA , Transcriptoma , Vertebrados/crescimento & desenvolvimento , Peixe-Zebra/embriologia
8.
Nat Methods ; 10(1): 77-83, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23202434

RESUMO

Lineage conversion of one somatic cell type to another is an attractive approach for generating specific human cell types. Lineage conversion can be direct, in the absence of proliferation and multipotent progenitor generation, or indirect, by the generation of expandable multipotent progenitor states. We report the development of a reprogramming methodology in which cells transition through a plastic intermediate state, induced by brief exposure to reprogramming factors, followed by differentiation. We use this approach to convert human fibroblasts to mesodermal progenitor cells, including by non-integrative approaches. These progenitor cells demonstrated bipotent differentiation potential and could generate endothelial and smooth muscle lineages. Differentiated endothelial cells exhibited neo-angiogenesis and anastomosis in vivo. This methodology for indirect lineage conversion to angioblast-like cells adds to the armamentarium of reprogramming approaches aimed at the study and treatment of ischemic pathologies.


Assuntos
Diferenciação Celular , Linhagem da Célula , Reprogramação Celular , Endotélio Vascular/citologia , Fibroblastos/citologia , Miócitos de Músculo Liso/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Western Blotting , Movimento Celular , Proliferação de Células , Células Cultivadas , Endotélio Vascular/metabolismo , Fibroblastos/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Camundongos , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo
9.
Methods Mol Biol ; 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38647861

RESUMO

Organoids derived from pluripotent stem cells exhibit notable similarities to organ development in vitro. Nonetheless, cardiac organoids generated to date possess immature phenotypes and are unable to model the full spectrum of heart development and disease. Here, we describe the developmental maturation of human heart organoids by controlled exposure to metabolic and hormonal factors over a 10-day period, mirroring key stages of human cardiac development and resulting in significant molecular, cellular, morphological, and functional changes. Overall, our findings represent a significant advancement in synthetic human heart development, offering a valuable platform for studying cardiac disease states and conducting pharmacological research.

10.
Artigo em Inglês | MEDLINE | ID: mdl-38830034

RESUMO

OBJECTIVES: Severe functional tricuspid regurgitation (FTR) is associated with subvalvular remodelling, but leaflet tissue alterations may also contribute. We set out to investigate molecular mechanisms driving leaflet remodelling in chronic ovine FTR. METHODS: Thirteen adult sheep (55 ± 4kg) underwent left thoracotomy, epicardial echocardiography, and pulmonary artery banding (PAB) to induce right heart failure and FTR. After 16 weeks, 13 banded (FTR) and 12 control (CTL) animals underwent median sternotomy for epicardial echocardiography and were subsequently sacrificed with each tricuspid leaflet tissue harvested for RNA-seq and histology. RESULTS: After 16 weeks, 7 animals developed severe, 2 moderate, and 4 mild tricuspid regurgitation (TR). Relative to CTL, FTR animals had increased PAP, TR, tricuspid annular diameter, and right atrial volume, while tricuspid annular plane systolic excursion (TAPSE) and RV fractional area change decreased. FTR leaflets exhibited altered constituents and an increase in cellularity. RNA-seq identified 85 significantly differentially expressed genes (DEG) with 17, 53, and 127 within the anterior, posterior, and septal leaflets respectively. RRM2, PRG4, and CXCL8 (IL-8) were identified as DEGs across all leaflets and CXCL8 was differentially expressed between FTR severity grades. RRM2, PRG4, and CXCL8 significantly correlated with TAPSE, and this correlation was consistent regardless of the anatomical location of the leaflet. CONCLUSIONS: PAB in our ovine model resulted in RV failure and FTR. Leaflet RNA-seq identified several DEGs, specifically RRM2, PRG4, and CXCL8, with known roles in tissue remodelling. These data along with an overall increase in leaflet cellularity suggest tricuspid leaflets actively remodel in FTR.

11.
Transplant Direct ; 10(7): e1658, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38881741

RESUMO

Background: Transplantation of human-induced pluripotent stem cell (hiPSC)-derived islet organoids is a promising cell replacement therapy for type 1 diabetes (T1D). It is important to improve the efficacy of islet organoids transplantation by identifying new transplantation sites with high vascularization and sufficient accommodation to support graft survival with a high capacity for oxygen delivery. Methods: A human-induced pluripotent stem cell line (hiPSCs-L1) was generated constitutively expressing luciferase. Luciferase-expressing hiPSCs were differentiated into islet organoids. The islet organoids were transplanted into the scapular brown adipose tissue (BAT) of nonobese diabetic/severe combined immunodeficiency disease (NOD/SCID) mice as the BAT group and under the left kidney capsule (KC) of NOD/SCID mice as a control group, respectively. Bioluminescence imaging (BLI) of the organoid grafts was performed on days 1, 7, 14, 28, 35, 42, 49, 56, and 63 posttransplantation. Results: BLI signals were detected in all recipients, including both the BAT and control groups. The BLI signal gradually decreased in both BAT and KC groups. However, the graft BLI signal intensity under the left KC decreased substantially faster than that of the BAT. Furthermore, our data show that islet organoids transplanted into streptozotocin-induced diabetic mice restored normoglycemia. Positron emission tomography/MRI verified that the islet organoids were transplanted at the intended location in these diabetic mice. Immunofluorescence staining revealed the presence of functional organoid grafts, as confirmed by insulin and glucagon staining. Conclusions: Our results demonstrate that BAT is a potentially desirable site for islet organoid transplantation for T1D therapy.

12.
Stem Cell Reports ; 19(3): 317-330, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38335962

RESUMO

Congenital heart defects are the most prevalent human birth defects, and their incidence is exacerbated by maternal health conditions, such as diabetes during the first trimester (pregestational diabetes). Our understanding of the pathology of these disorders is hindered by a lack of human models and the inaccessibility of embryonic tissue. Using an advanced human heart organoid system, we simulated embryonic heart development under pregestational diabetes-like conditions. These organoids developed pathophysiological features observed in mouse and human studies before, including ROS-mediated stress and cardiomyocyte hypertrophy. scRNA-seq revealed cardiac cell-type-specific dysfunction affecting epicardial and cardiomyocyte populations and alterations in the endoplasmic reticulum and very-long-chain fatty acid lipid metabolism. Imaging and lipidomics confirmed these findings and showed that dyslipidemia was linked to fatty acid desaturase 2 mRNA decay dependent on IRE1-RIDD signaling. Targeting IRE1 or restoring lipid levels partially reversed the effects of pregestational diabetes, offering potential preventive and therapeutic strategies in humans.


Assuntos
Cardiomiopatias , Diabetes Mellitus , Cardiopatias Congênitas , Humanos , Camundongos , Animais , Cardiopatias Congênitas/patologia , Estresse do Retículo Endoplasmático/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Organoides/metabolismo , Lipídeos
13.
Front Bioeng Biotechnol ; 11: 1214431, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37560538

RESUMO

In recent years, significant biotechnological advancements have been made in engineering human cardiac tissues and organ-like models. This field of research is crucial for both basic and translational research due to cardiovascular disease being the leading cause of death in the developed world. Additionally, drug-associated cardiotoxicity poses a major challenge for drug development in the pharmaceutical and biotechnological industries. Progress in three-dimensional cell culture and microfluidic devices has enabled the generation of human cardiac models that faithfully recapitulate key aspects of human physiology. In this review, we will discuss 3D pluripotent stem cell (PSC)-models of the human heart, such as engineered heart tissues and organoids, and their applications in disease modeling and drug screening.

14.
Methods Mol Biol ; 2592: 195-206, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36507995

RESUMO

Pancreatic islet transplantation is a promising cell replacement treatment for patients afflicted with type 1 diabetes (T1D), which is an autoimmune disease resulting in the destruction of insulin-producing islet ß-cells. However, the shortage of donor pancreatic islets significantly hampers the widespread application of this strategy as routine therapy. Pluripotent stem cell-derived insulin-producing islet organoids constitute a promising alternative ß-cell source for T1D patients. Early after transplantation, it is critical to know the fate of transplanted islet organoids, but determining their survival remains a significant technical challenge. Bioluminescence imaging (BLI) is an optical molecular imaging technique that detects the survival of living cells using light emitted from luciferase-expressing bioreporter cells. Through BLI, the post-transplantation fate of islet organoids can be evaluated over time in a noninvasive fashion with minimal intervention, thus making BLI an ideal tool to determine the success of the transplant and improving cell replacement therapy approaches for T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Humanos , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Organoides/metabolismo , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 1/metabolismo , Insulina/metabolismo
15.
Sci Rep ; 13(1): 18046, 2023 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-37872254

RESUMO

Three-hourly CMIP6 projections have been used in conjuction with the CSIRO WaveWatchIII wave model to calculate the global trends in offshore wind and wave energy for the SSP585 and SSP126 scenarios until 2100. The results indicate that moderate yet significant changes are expected in the theoretical electricity generated from wind and waves at fewer than 10-15% of coastal locations. While this implies a generally stable outlook for the future, certain coastal regions with existing or planned wind farms may experience a slight reduction in production by 2100. Regarding wave energy, given its early stage of development, a more cautious approach is advisable, although a similar conclusion may be reached. Considering the decreasing installation costs on the horizon and accounting for both climatic scenarios, this provides a reliable context for most ongoing feasibility studies, technological developments, and offshore facility investments.

16.
bioRxiv ; 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37333095

RESUMO

Congenital heart defects constitute the most common birth defect in humans, affecting approximately 1% of all live births. The incidence of congenital heart defects is exacerbated by maternal conditions, such as diabetes during the first trimester. Our ability to mechanistically understand these disorders is severely limited by the lack of human models and the inaccessibility to human tissue at relevant stages. Here, we used an advanced human heart organoid model that recapitulates complex aspects of heart development during the first trimester to model the effects of pregestational diabetes in the human embryonic heart. We observed that heart organoids in diabetic conditions develop pathophysiological hallmarks like those previously reported in mouse and human studies, including ROS-mediated stress and cardiomyocyte hypertrophy, among others. Single cell RNA-seq revealed cardiac cell type specific-dysfunction affecting epicardial and cardiomyocyte populations, and suggested alterations in endoplasmic reticulum function and very long chain fatty acid lipid metabolism. Confocal imaging and LC-MS lipidomics confirmed our observations and showed that dyslipidemia was mediated by fatty acid desaturase 2 (FADS2) mRNA decay dependent on IRE1-RIDD signaling. We also found that the effects of pregestational diabetes could be reversed to a significant extent using drug interventions targeting either IRE1 or restoring healthy lipid levels within organoids, opening the door to new preventative and therapeutic strategies in humans.

17.
Nat Commun ; 14(1): 8245, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38086920

RESUMO

Pluripotent stem cell-derived organoids can recapitulate significant features of organ development in vitro. We hypothesized that creating human heart organoids by mimicking aspects of in utero gestation (e.g., addition of metabolic and hormonal factors) would lead to higher physiological and anatomical relevance. We find that heart organoids produced using this self-organization-driven developmental induction strategy are remarkably similar transcriptionally and morphologically to age-matched human embryonic hearts. We also show that they recapitulate several aspects of cardiac development, including large atrial and ventricular chambers, proepicardial organ formation, and retinoic acid-mediated anterior-posterior patterning, mimicking the developmental processes found in the post-heart tube stage primitive heart. Moreover, we provide proof-of-concept demonstration of the value of this system for disease modeling by exploring the effects of ondansetron, a drug administered to pregnant women and associated with congenital heart defects. These findings constitute a significant technical advance for synthetic heart development and provide a powerful tool for cardiac disease modeling.


Assuntos
Cardiopatias , Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Gravidez , Humanos , Feminino , Células-Tronco Pluripotentes Induzidas/metabolismo , Organoides/metabolismo , Coração , Cardiopatias/metabolismo , Diferenciação Celular/fisiologia
18.
Eur Cell Mater ; 24: 90-106; discussion 106, 2012 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-22828988

RESUMO

Smart biomaterials play a key role when aiming at successful tissue repair by means of regenerative medicine approaches, and are expected to contain chemical as well as mechanical cues that will guide the regenerative process. Recent advances in the understanding of stem cell biology and mechanosensing have shed new light onto the importance of the local microenvironment in determining cell fate. Herein we report the biological properties of a bioactive, biodegradable calcium phosphate glass/polylactic acid composite biomaterial that promotes bone marrow-derived endothelial progenitor cell (EPC) mobilisation, differentiation and angiogenesis through the creation of a controlled bone healing-like microenvironment. The angiogenic response is triggered by biochemical and mechanical cues provided by the composite, which activate two synergistic cell signalling pathways: a biochemical one mediated by the calcium-sensing receptor and a mechanosensitive one regulated by non-muscle myosin II contraction. Together, these signals promote a synergistic response by activating EPCs-mediated VEGF and VEGFR-2 synthesis, which in turn promote progenitor cell homing, differentiation and tubulogenesis. These findings highlight the importance of controlling microenvironmental cues for stem/progenitor cell tissue engineering and offer exciting new therapeutical opportunities for biomaterial-based vascularisation approaches and clinical applications.


Assuntos
Materiais Biocompatíveis/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Células Endoteliais/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Materiais Biocompatíveis/química , Medula Óssea/efeitos dos fármacos , Fosfatos de Cálcio/química , Sinalização do Cálcio/fisiologia , Diferenciação Celular/efeitos dos fármacos , Microambiente Celular , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Vidro/química , Ácido Láctico/química , Mecanotransdução Celular/fisiologia , Miosina Tipo II/metabolismo , Poliésteres , Polímeros/química , Ratos , Ratos Endogâmicos Lew , Receptores de Detecção de Cálcio/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Engenharia Tecidual , Alicerces Teciduais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Curr Protoc ; 2(6): e461, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35723517

RESUMO

Congenital heart defects (CHD) constitute the most common type of birth defect in humans. Maternal diabetes during the first trimester of pregnancy (pregestational diabetes, or PGD) is one of the most prominent factors contributing to CHD, and is present in a significant population of female patients with diabetes in reproductive age. PGD is challenging to manage clinically due to the extreme sensitivity of the developing embryo to glucose oscillations, and constitutes a critical health problem for the mother and the fetus. The prevalence of PGD-induced CHD is increasing due to the ongoing diabetes epidemic. While studies using animal models and cells in culture have demonstrated that PGD alters critical cellular and developmental processes, the mechanisms remain obscure, and it is unclear to what extent these models recapitulate PGD-induced CHD in humans. Clinical practice precludes direct studies in developing human embryos, further highlighting the need for physiologically relevant models. To bypass many of these technical and ethical limitations, we describe here a human pluripotent stem cell (hPSC)-based method to generate developmentally relevant self-organizing human heart organoids. By using glucose and insulin to mimic the diabetic environment that the embryo faces in PGD, this system allows modeling critical features of PGD in a human system with relevant physiology, structure, and cell types. The protocol starts with the generation of hPSC-derived embryoid bodies in a 96-well plate, followed by a small molecule-based three-step Wnt activation/inhibition/activation strategy. Organoids are then differentiated under healthy (normal insulin and glucose) and diabetic conditions (high insulin and glucose) over time, allowing for the study of the effects of pregestational diabetes on the developing human heart. We also provide an immunofluorescence protocol for comparing, characterizing, and analyzing the differences between the healthy and diabetic organoids, and comment on additional steps for preparing the organoids for analysis by other techniques after differentiation. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Generation of hPSC-derived embryoid bodies Basic Protocol 2: Differentiation of EBs into heart organoids under healthy and diabetes-like conditions Basic Protocol 3: Immunofluorescence and organoid preparation for other assays.


Assuntos
Diabetes Mellitus , Células-Tronco Pluripotentes , Diabetes Mellitus/metabolismo , Feminino , Glucose/metabolismo , Humanos , Insulina/metabolismo , Organoides
20.
Biomater Adv ; 139: 213035, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35907761

RESUMO

In situ tissue engineering strategies are a promising approach to activate the endogenous regenerative potential of the cardiac tissue helping the heart to heal itself after an injury. However, the current use of complex reprogramming vectors for the activation of reparative pathways challenges the easy translation of these therapies into the clinic. Here, we evaluated the response of mouse neonatal and human induced pluripotent stem cell-derived cardiomyocytes to the presence of exogenous lactate, thus mimicking the metabolic environment of the fetal heart. An increase in cardiomyocyte cell cycle activity was observed in the presence of lactate, as determined through Ki67 and Aurora-B kinase. Gene expression and RNA-sequencing data revealed that cardiomyocytes incubated with lactate showed upregulation of BMP10, LIN28 or TCIM in tandem with downregulation of GRIK1 or DGKK among others. Lactate also demonstrated a capability to modulate the production of inflammatory cytokines on cardiac fibroblasts, reducing the production of Fas, Fraktalkine or IL-12p40, while stimulating IL-13 and SDF1a. In addition, the generation of a lactate-rich environment improved ex vivo neonatal heart culture, by affecting the contractile activity and sarcomeric structures and inhibiting epicardial cell spreading. Our results also suggested a common link between the effect of lactate and the activation of hypoxia signaling pathways. These findings support a novel use of lactate in cardiac tissue engineering, modulating the metabolic environment of the heart and thus paving the way to the development of lactate-releasing platforms for in situ cardiac regeneration.


Assuntos
Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Ácido Láctico/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA