Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Exp Cell Res ; 436(1): 113961, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38341080

RESUMEN

Non-coding RNAs, particularly small Cajal-body associated RNAs (scaRNAs), play a significant role in spliceosomal RNA modifications. While their involvement in ischemic myocardium regeneration is known, their role in cardiac development is unexplored. We investigated scaRNA20's role in iPSC differentiation into cardiomyocytes (iCMCs) via overexpression and knockdown assays. We measured scaRNA20-OE-iCMCs and scaRNA20-KD-iCMCs contractility using Particle Image Velocimetry (PIV), comparing them to control iCMCs. We explored scaRNA20's impact on alternative splicing via pseudouridylation (Ψ) of snRNA U12, analyzing its functional consequences in cardiac differentiation. scaRNA20-OE-iPSC differentiation increased beating colonies, upregulated cardiac-specific genes, activated TP53 and STAT3, and preserved contractility under hypoxia. Conversely, scaRNA20-KD-iCMCs exhibited poor differentiation and contractility. STAT3 inhibition in scaRNA20-OE-iPSCs hindered cardiac differentiation. RNA immunoprecipitation revealed increased Ψ at the 28th uridine of U12 RNA in scaRNA20-OE iCMCs. U12-KD iCMCs had reduced cardiac differentiation, which improved upon U12 RNA introduction. In summary, scaRNA20-OE in iPSCs enhances cardiomyogenesis, preserves iCMC function under hypoxia, and may have implications for ischemic myocardium regeneration.


Asunto(s)
ARN Nuclear Pequeño , ARN , Humanos , ARN Nuclear Pequeño/genética , Empalme Alternativo , Hipoxia , Miocitos Cardíacos
2.
Pflugers Arch ; 474(1): 63-81, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34967935

RESUMEN

Experimental and clinical evidence suggests that diabetic subjects are predisposed to a distinct cardiovascular dysfunction, known as diabetic cardiomyopathy (DCM), which could be an autonomous disease independent of concomitant micro and macrovascular disorders. DCM is one of the prominent causes of global morbidity and mortality and is on a rising trend with the increase in the prevalence of diabetes mellitus (DM). DCM is characterized by an early left ventricle diastolic dysfunction associated with the slow progression of cardiomyocyte hypertrophy leading to heart failure, which still has no effective therapy. Although the well-known "Renin Angiotensin Aldosterone System (RAAS)" inhibition is considered a gold-standard treatment in heart failure, its role in DCM is still unclear. At the cellular level of DCM, RAAS induces various secondary mechanisms, adding complications to poor prognosis and treatment of DCM. This review highlights the importance of RAAS signaling and its major secondary mechanisms involving inflammation, oxidative stress, mitochondrial dysfunction, and autophagy, their role in establishing DCM. In addition, studies lacking in the specific area of DCM are also highlighted. Therefore, understanding the complex role of RAAS in DCM may lead to the identification of better prognosis and therapeutic strategies in treating DCM.


Asunto(s)
Cardiomiopatías Diabéticas/etiología , Sistema Renina-Angiotensina , Angiotensina II/metabolismo , Animales , Autofagia , Cardiomiopatías Diabéticas/metabolismo , Humanos , Inflamación/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Estrés Oxidativo , Peptidil-Dipeptidasa A/metabolismo , Receptores de Angiotensina/metabolismo
3.
Exp Cell Res ; 400(1): 112508, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33549576

RESUMEN

Noonan syndrome (NS) is a dominant autosomal genetic disorder, associated with mutations in several genes that exhibit multisystem abnormal development including cardiac defects. NS associated with the Son of Sevenless homolog 1 (SOS1) gene mutation attributes to the development of cardiomyopathy and congenital heart defects. Since the treatment option for NS is very limited, an in vitro disease model with SOS1 gene mutation would be beneficial for exploring therapeutic possibilities for NS. We reprogrammed cardiac fibroblasts obtained from a NS patient and normal control skin fibroblasts (C-SF) into induced pluripotent stem cells (iPSCs). We identified NS-iPSCs carry a heterozygous single nucleotide variation in the SOS1 gene at the c.1654A > G. Furthermore, the control and NS-iPSCs were differentiated into induced cardiomyocytes (iCMCs), and the electron microscopic analysis showed that the sarcomeres of the NS-iCMCs were highly disorganized. FACS analysis showed that 47.5% of the NS-iCMCs co-expressed GATA4 and cardiac troponin T proteins, and the mRNA expression levels of many cardiac related genes, studied by qRT-PCR array, were significantly reduced when compared to the control C-iCMCs. We report for the first time that NS-iPSCs carry a single nucleotide variation in the SOS1 gene at the c.1654A>G were showing significantly reduced cardiac genes and proteins expression as well as structurally and functionally compromised when compared to C-iCMCs. These iPSCs and iCMCs can be used as a modeling platform to unravel the pathologic mechanisms and also the development of novel drug for the cardiomyopathy in patients with NS.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Mutación , Miocitos Cardíacos/patología , Síndrome de Noonan/patología , Proteína SOS1/genética , Estudios de Casos y Controles , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Síndrome de Noonan/genética
4.
J Cell Mol Med ; 25(18): 8904-8919, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34390186

RESUMEN

Generation of induced pluripotent stem cells (iPSCs) and their differentiation into mesenchymal stem/stromal cells (iMSCs) have created exciting source of cells for autologous therapy. In this study, we have compared the therapeutic potential of iMSCs generated from urinary epithelial (UE) cells with the available umbilical cord MSCs (UC-MSCs). For this, adult UE cells were treated with the mRNA of pluripotent genes (OCT4, NANOG, SOX2, KLF4, MYC and LIN28) and a cocktail of miRNAs under specific culture conditions for generating iPSCs. Our non-viral and mRNA-based treatment regimen demonstrated a high reprogramming efficiency to about 30% at passage 0. These UE-iPSCs were successfully differentiated further into ectoderm, endoderm and mesoderm lineage of cells. Moreover, these UE-iPSCs were subsequently differentiated into iMSCs and were compared with the UC-MSCs. These iMSCs were capable of differentiating into osteocytes, chondrocytes and adipocytes. Our qRT-PCR and Western blot data showed that the CD73, CD90 and CD105 gene transcripts and proteins were highly expressed in iMSCs and UC-MSCs but not in other cells. The comparative qRT-PCR data showed that the iMSCs maintained their MSC characteristics without any chromosomal abnormalities even at later passages (P15), during which the UC-MSCs started losing their MSC characteristics. Importantly, the wound-healing property demonstrated through migration assay was superior in iMSCs when compared to the UC-MSCs. In this study, we have demonstrated an excellent non-invasive and pain-free method of obtaining iMSCs for regenerative therapy. These homogeneous autologous highly proliferative iMSCs may provide an alternative source of cells to UC-MSCs for treating various diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Medicina Regenerativa/métodos , Cordón Umbilical/citología , Diferenciación Celular , Células Cultivadas , Humanos , Masculino , Persona de Mediana Edad
5.
Curr Genomics ; 22(7): 526-540, 2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-35386434

RESUMEN

Aims: Perform in-silico analysis of human SOS1 mutations to elucidate their pathogenic role in Noonan syndrome (NS). Background: NS is an autosomal dominant genetic disorder caused by single nucleotide mutation in PTPN11, SOS1, RAF1, and KRAS genes. NS is thought to affect approximately 1 in 1000. NS patients suffer different pathogenic effects depending on the mutations they carry. Analysis of the mutations would be a promising predictor in identifying the pathogenic effect of NS. Methods: We performed computational analysis of the SOS1 gene to identify the pathogenic nonsynonymous single nucleotide polymorphisms (nsSNPs) th a t cause NS. SOS1 variants were retrieved from the SNP database (dbSNP) and analyzed by in-silico tools I-Mutant, iPTREESTAB, and MutPred to elucidate their structural and functional characteristics. Results: We found that 11 nsSNPs of SOS1 that were linked to NS. 3D modeling of the wild-type and the 11 nsSNPs of SOS1 showed that SOS1 interacts with cardiac proteins GATA4, TNNT2, and ACTN2. We also found that GRB2 and HRAS act as intermediate molecules between SOS1 and cardiac proteins. Our in-silico analysis findings were further validated using induced cardiomyocytes (iCMCs) derived from NS patients carrying SOS1 gene variant c.1654A>G (NSiCMCs) and compared to control human skin fibroblast-derived iCMCs (C-iCMCs). Our in vitro data confirmed that the SOS1, GRB2 and HRAS gene expressions as well as the activated ERK protein, were significantly decreased in NS-iCMCs when compared to C-iCMCs. Conclusion: This is the first in-silico and in vitro study demonstrating that 11 nsSNPs of SOS1 play deleterious pathogenic roles in causing NS.

6.
Differentiation ; 112: 58-66, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31954271

RESUMEN

Despite significant effort devoted to developing new treatments and procedures, cardiac disease is still one of the leading causes of death in the world. The loss of myocytes due to ischemic injury remains a major therapeutic challenge. However, cell-based therapy to repair the injured heart has shown significant promise in basic and translation research and in clinical trials. Embryonic stem cells have been successfully used to improve cardiac outcomes. Unfortunately, treatment with these cells is complicated by ethical and legal issues. Recent progress in developing induced pluripotent stem cells (iPSCs) using non-viral vectors has made it possible to derive cardiomyocytes for therapy. This review will focus on these non-integration-based approaches for reprogramming and their therapeutic advantages for cardiovascular medicine.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Cardiopatías/terapia , Células Madre Pluripotentes Inducidas/trasplante , Miocitos Cardíacos/trasplante , Diferenciación Celular/genética , Reprogramación Celular/genética , Vectores Genéticos/uso terapéutico , Humanos , Medicina Regenerativa/tendencias
7.
Molecules ; 25(2)2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31968561

RESUMEN

Kleinia pendula (Forssk.) DC. is a prostrate or pendent dark green succulent herb found in the southwestern mountain regions of Saudi Arabia. The literature survey of the plant reveals a lack of phytochemical and pharmacological studies, although traditional uses have been noted. The objective of the present work was to assess the in vivo analgesic and anti-inflammatory activities, as well as, the in vitro cytotoxic potential of the fractions of Kleinia pendula, and correlate these activities to the plant metabolites. The methanolic extract of Kleinia pendula was subjected to fractionation with n-hexane, ethyl acetate, chloroform, n-butanol, and water. The fractions were screened for their analgesic and anti-inflammatory activities, as well as cytotoxic activity against breast, liver, and colon cancer cell lines. The n-hexane and chloroform fractions of Kleinia pendula showed significant cytotoxic activity against all three cancer cell lines tested. The ethyl acetate and chloroform fractions showed significant analgesic and anti-inflammatory activities. The metabolites in these three active fractions were determined using UPLC-PDA-ESI-MS. Thus, the analgesic and anti-inflammatory activities of the plant were attributed to its phenolic acids (caffeoylquinic acid derivatives, protocatechuic, and chlorogenic acids). While fatty acids and triterpenoids such as (tormentic acid) in the hexane fraction are responsible for the cytotoxic activity; thus, these fractions of Kleinia pendula may be a novel source for the development of new plant-based analgesic, anti-inflammatory, and anticancer drugs.


Asunto(s)
Analgésicos/farmacología , Antiinflamatorios/farmacología , Antineoplásicos Fitogénicos/farmacología , Fabaceae/química , Fitoquímicos/farmacología , Analgésicos/química , Antiinflamatorios/química , Antineoplásicos Fitogénicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Células HCT116 , Células Hep G2 , Humanos , Células MCF-7 , Espectrometría de Masas , Fitoquímicos/química , Extractos Vegetales/química , Arabia Saudita , Senecio
8.
Cells ; 13(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38786076

RESUMEN

Cardiovascular diseases continue to challenge global health, demanding innovative therapeutic solutions. This review delves into the transformative role of mesenchymal stem cells (MSCs) in advancing cardiovascular therapeutics. Beginning with a historical perspective, we trace the development of stem cell research related to cardiovascular diseases, highlighting foundational therapeutic approaches and the evolution of cell-based treatments. Recognizing the inherent challenges of MSC-based cardiovascular therapeutics, which range from understanding the pro-reparative activity of MSCs to tailoring patient-specific treatments, we emphasize the need to refine the pro-regenerative capacity of these cells. Crucially, our focus then shifts to the strategies of the fourth generation of cell-based therapies: leveraging the secretomic prowess of MSCs, particularly the role of extracellular vesicles; integrating biocompatible scaffolds and artificial sheets to amplify MSCs' potential; adopting three-dimensional ex vivo propagation tailored to specific tissue niches; harnessing the promise of genetic modifications for targeted tissue repair; and institutionalizing good manufacturing practice protocols to ensure therapeutic safety and efficacy. We conclude with reflections on these advancements, envisaging a future landscape redefined by MSCs in cardiovascular regeneration. This review offers both a consolidation of our current understanding and a view toward imminent therapeutic horizons.


Asunto(s)
Enfermedades Cardiovasculares , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/citología , Enfermedades Cardiovasculares/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Tratamiento Basado en Trasplante de Células y Tejidos/métodos
9.
Cells ; 13(6)2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38534355

RESUMEN

Neuroinflammatory and neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI) and Amyotrophic lateral sclerosis (ALS) are chronic major health disorders. The exact mechanism of the neuroimmune dysfunctions of these disease pathogeneses is currently not clearly understood. These disorders show dysregulated neuroimmune and inflammatory responses, including activation of neurons, glial cells, and neurovascular unit damage associated with excessive release of proinflammatory cytokines, chemokines, neurotoxic mediators, and infiltration of peripheral immune cells into the brain, as well as entry of inflammatory mediators through damaged neurovascular endothelial cells, blood-brain barrier and tight junction proteins. Activation of glial cells and immune cells leads to the release of many inflammatory and neurotoxic molecules that cause neuroinflammation and neurodegeneration. Gulf War Illness (GWI) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are chronic disorders that are also associated with neuroimmune dysfunctions. Currently, there are no effective disease-modifying therapeutic options available for these diseases. Human induced pluripotent stem cell (iPSC)-derived neurons, astrocytes, microglia, endothelial cells and pericytes are currently used for many disease models for drug discovery. This review highlights certain recent trends in neuroinflammatory responses and iPSC-derived brain cell applications in neuroinflammatory disorders.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas , Humanos , Enfermedades Neuroinflamatorias , Células Endoteliales , Inflamación
10.
Exp Biol Med (Maywood) ; 248(5): 434-444, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36740769

RESUMEN

Exosomes are extracellular vesicles released by many cell types with varying compositions. Major bioactive factors present in exosomes are protein, lipid, mRNA, and miRNA. Exosomes are fundamental regulators of cellular trafficking and signaling in both physiological and pathological conditions. Various conditions such as oxidative stress, endoplasmic reticulum stress, ribosomal stress, and thermal stress alter the concentration of exosomal mRNA, and miRNA, lipids, and proteins. Stem cell-derived exosomes have been shown to regulate a variety of stresses, either inhibiting or promoting cell balance. Stem cell-derived exosomes direct the crosstalk between various cell types which helps recovery by transferring information in proteins, lipids, and so on. This is one of the reasons why exosomes are used as biomarkers for a multitude of disease conditions. This review highlights the bioengineering of fabricated exosomal cargoes. It includes the manipulation and delivery of specific exosomal cargoes such as noncoding RNAs, recombinant proteins, immune modulators, therapeutic drugs, and small molecules. Such therapeutic approaches may precisely deliver the therapeutic drugs at the target site in the management of various disease conditions. Importantly, we have focused on the therapeutic applications of stem cell-derived exosomes in cardiovascular disease conditions such as myocardial infarction, ischemic heart disease, cardiomyopathy, heart failure, sepsis, and cardiac fibrosis. Generally, two approaches are being followed by researchers for exosomal bioengineering. This literature review will shed light on the role of stem cell-derived exosomes in stress balance and provides a new avenue for the treatment of cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Exosomas , MicroARNs , Humanos , Enfermedades Cardiovasculares/terapia , Exosomas/metabolismo , MicroARNs/genética , Células Madre/metabolismo , ARN Mensajero/metabolismo , Lípidos
11.
Cells ; 12(18)2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37759491

RESUMEN

Cardiac injury, such as myocardial infarction and heart failure, remains a significant global health burden. The limited regenerative capacity of the adult heart poses a challenge for restoring its function after injury. Mesenchymal stem cells (MSCs) have emerged as promising candidates for cardiac regeneration due to their ability to differentiate into various cell types and secrete bioactive molecules. In recent years, attention has been given to noncoding RNAs derived from MSCs, particularly long noncoding RNAs (lncRNAs), and their potential role in cardiac injury and repair. LncRNAs are RNA molecules that do not encode proteins but play critical roles in gene regulation and cellular responses including cardiac repair and regeneration. This review focused on MSC-derived lncRNAs and their implications in cardiac regeneration, including their effects on cardiac function, myocardial remodeling, cardiomyocyte injury, and angiogenesis. Understanding the molecular mechanisms of MSC-derived lncRNAs in cardiac injury and repair may contribute to the development of novel therapeutic strategies for treating cardiovascular diseases. However, further research is needed to fully elucidate the potential of MSC-derived lncRNAs and address the challenges in this field.


Asunto(s)
Lesiones Cardíacas , Células Madre Mesenquimatosas , Infarto del Miocardio , ARN Largo no Codificante , Adulto , Humanos , ARN Largo no Codificante/genética , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Miocitos Cardíacos
12.
Cells ; 12(8)2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37190068

RESUMEN

Stem cells' self-renewal and multi-lineage differentiation are regulated by a complex network consisting of signaling factors, chromatin regulators, transcription factors, and non-coding RNAs (ncRNAs). Diverse role of ncRNAs in stem cell development and maintenance of bone homeostasis have been discovered recently. The ncRNAs, such as long non-coding RNAs, micro RNAs, circular RNAs, small interfering RNA, Piwi-interacting RNAs, etc., are not translated into proteins but act as essential epigenetic regulators in stem cells' self-renewal and differentiation. Different signaling pathways are monitored efficiently by the differential expression of ncRNAs, which function as regulatory elements in determining the fate of stem cells. In addition, several species of ncRNAs could serve as potential molecular biomarkers in early diagnosis of bone diseases, including osteoporosis, osteoarthritis, and bone cancers, ultimately leading to the development of new therapeutic strategies. This review aims to explore the specific roles of ncRNAs and their effective molecular mechanisms in the growth and development of stem cells, and in the regulation of osteoblast and osteoclast activities. Furthermore, we focus on and explore the association of altered ncRNA expression with stem cells and bone turnover.


Asunto(s)
Enfermedades Óseas , MicroARNs , ARN Largo no Codificante , Humanos , ARN no Traducido/genética , ARN no Traducido/metabolismo , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Diferenciación Celular/genética , Enfermedades Óseas/genética , Enfermedades Óseas/terapia
13.
J Cell Mol Med ; 16(1): 174-84, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21352470

RESUMEN

The major problem in stem cell therapy includes viability and engraftment efficacy of stem cells after transplantation. Indeed, the vast majority of host-transfused cells do not survive beyond 24-72 hrs. To increase the survival and engraftment of implanted cardiac stem cells in the host, we developed a technique of treating these cells with resveratrol, and tested it in a rat model of left anterior descending (LAD) occlusion. Multi-potent clonogenic cardiac stem cells isolated from rat heart and stably transfected with EGFP were pre-treated with 2.5 µM resveratrol for 60 min. Rats were anaesthetized, hearts opened and the LAD occluded to induce heart attack. One week later, the cardiac reduced environment was confirmed in resveratrol treated rat hearts by the enhanced expression of nuclear factor-E2-related factor-2 (Nrf2) and redox effector factor-1 (Ref-1). M-mode echocardiography after stem cell therapy, showed improvement in cardiac function (left ventricular ejection fraction, fractional shortening and cardiac output) in both, the treated and control group after 7 days, but only resveratrol-modified stem cell group revealed improvement in cardiac function at the end of 1, 2 and 4 months time. The improvement of cardiac function was accompanied by enhanced stem cell survival and engraftment as demonstrated by the expression of cell proliferation marker Ki67 and differentiation of stem cells towards the regeneration of the myocardium as demonstrated by the expression of EGFP up to 4 months after LAD occlusion in the resveratrol-treated stem cell group. Expression of stromal cell-derived factor and myosin conclusively demonstrated homing of stem cells in the infarcted myocardium, its regeneration leading to improvement of cardiac function.


Asunto(s)
Infarto del Miocardio/terapia , Miocardio/citología , Regeneración/fisiología , Trasplante de Células Madre/métodos , Células Madre/efectos de los fármacos , Células Madre/fisiología , Estilbenos/farmacología , Animales , Antioxidantes/farmacología , Biomarcadores/metabolismo , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Ecocardiografía , Corazón/fisiología , Masculino , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Resveratrol , Células Madre/citología
14.
Mol Cell Biochem ; 360(1-2): 35-45, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21918828

RESUMEN

Most clinical trials with vitamin E could not lower cholesterol and thus, have been deemed unsuccessful. Recently, tocotrienols, isomers of vitamin E have been found to lower LDL levels. To explore if tocotrienols could be the drug target for vitamin E, rabbits were kept on cholesterol diet for 60 days supplemented with tocotrienol-α, tocotrienol-δ, and tocotrienol-γ for the last 30 days. The serum cholesterol levels (in mmol/l) were 24.4 (tocotrienol-α), 34.9 (tocotrienol-δ), 19.8 (tocotrienol-γ) vs. 39.7 (control). Left ventricular function including aortic flow and developed pressure exhibited significantly improved recovery with tocotrienol-γ and -α, but not with tocotrienol-δ. The myocardial infarct size showed a similar pattern: 33% (tocotrienol-α), 23% (tocotrienol-γ), and 47% (tocotrienol-δ). To examine the molecular mechanisms of cardioprotective effects, gene expression profile was determined using Atlas 1.2/1.2II followed by determination of gene profiles using PedQuest 8.3 software. Based on genomic profiles, the following cholesterol-related proteins were examined: FABP, TGF-ß (cholesterol suppresses TGF-ß), ET-1 (increased by hypercholesterolemia), SPOT 14 (linked with hypercholesterolemia), and matrix metalloproteinase (MMP) 2 and MMP9 (cholesterol regulates MMP2 and MMP9 expression) in the heart. Consistent with the cardioprotective effects of tocotrienol-α and -γ, these two isomers reduced ET-1, decreased MMP2 and MM9, increased TGF-ß and reduced SPOT 14, while tocotrienol-δ had no effects. The results of the present study demonstrate that the two isomers of tocotrienols, α and γ, render the hypercholesterolemic hearts resistant to ischemic reperfusion injury by lowering several hypercholesterolemic proteins including MMP2, MMP9, ET-1, and SPOT 14 and upregulating TGF-ß.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Cardiotónicos/uso terapéutico , Cromanos/uso terapéutico , Hipercolesterolemia/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Vitamina E/análogos & derivados , Animales , Aterosclerosis/etiología , Aterosclerosis/prevención & control , Colesterol/sangre , Dieta Alta en Grasa , Endotelina-1/genética , Endotelina-1/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Corazón/efectos de los fármacos , Hipercolesterolemia/sangre , Hipercolesterolemia/complicaciones , Técnicas In Vitro , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Conejos , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Factores Sexuales , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Vitamina E/uso terapéutico
15.
Circ Res ; 107(11): 1374-86, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-21088285

RESUMEN

RATIONALE: The turnover of cardiomyocytes in the aging female and male heart is currently unknown, emphasizing the need to define human myocardial biology. OBJECTIVE: The effects of age and gender on the magnitude of myocyte regeneration and the origin of newly formed cardiomyocytes were determined. METHODS AND RESULTS: The interaction of myocyte replacement, cellular senescence, growth inhibition, and apoptosis was measured in normal female (n=32) and male (n=42) human hearts collected from patients 19 to 104 years of age who died from causes other than cardiovascular diseases. A progressive loss of telomeric DNA in human cardiac stem cells (hCSCs) occurs with aging and the newly formed cardiomyocytes inherit short telomeres and rapidly reach the senescent phenotype. Our data provide novel information on the superior ability of the female heart to sustain the multiple variables associated with the development of the senescent myopathy. At all ages, the female heart is equipped with a larger pool of functionally competent hCSCs and younger myocytes than the male myocardium. The replicative potential is higher and telomeres are longer in female hCSCs than in male hCSCs. In the female heart, myocyte turnover occurs at a rate of 10%, 14%, and 40% per year at 20, 60, and 100 years of age, respectively. Corresponding values in the male heart are 7%, 12%, and 32% per year, documenting that cardiomyogenesis involves a large and progressively increasing number of parenchymal cells with aging. From 20 to 100 years of age, the myocyte compartment is replaced 15 times in women and 11 times in men. CONCLUSIONS: The human heart is a highly dynamic organ regulated by a pool of resident hCSCs that modulate cardiac homeostasis and condition organ aging.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Diferenciación Celular/fisiología , Senescencia Celular/fisiología , Corazón/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/fisiología , Muerte Celular/fisiología , Células Cultivadas , Femenino , Corazón/anatomía & histología , Humanos , Masculino , Persona de Mediana Edad , Caracteres Sexuales , Adulto Joven
16.
Biometals ; 25(1): 203-17, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22089858

RESUMEN

Although iron is known to be a component of the pathogenesis and/or maintenance of acute lung injury (ALI) in experimental animals and human subjects, the majority of these studies have focused on disturbances in iron homeostasis in the airways resulting from exposure to noxious gases and particles. Considerably less is known about the effect of increased plasma levels of redox-reactive non-transferrin bound iron (NTBI) and its impact on pulmonary endothelium. Plasma levels of NTBI can increase under various pathophysiological conditions, including those associated with ALI, and multiple mechanisms are in place to affect the [Fe(2+)]/[Fe(3+)] redox steady state. It is well accepted, however, that intracellular transport of NTBI occurs after reduction of [Fe(3+)] to [Fe(2+)] (and is mediated by divalent metal transporters). Accordingly, as an experimental model to investigate mechanisms mediating vascular effects of redox reactive iron, rat pulmonary artery endothelial cells (RPAECs) were subjected to pulse treatment (10 min) with [Fe(2+)] nitriloacetate (30 µM) in the presence of pyrithione, an iron ionophore, to acutely increase intracellular labile pool of iron. Cellular iron influx and cell shape profile were monitored with time-lapse imaging techniques. Exposure of RPAECs to [Fe(2+)] resulted in: (i) an increase in intracellular iron as detected by the iron sensitive fluorophore, PhenGreen; (ii) depletion of cell glutathione; and (iii) nuclear translocation of stress-response transcriptional factors Nrf2 and NFkB (p65). The resulting iron-induced cell alterations were characterized by cell polarization and formation of membrane cuplike and microvilli-like projections abundant with ICAM-1, caveolin-1, and F-actin. The iron-induced re-arrangements in cytoskeleton, alterations in focal cell-cell interactions, and cell buckling were accompanied by decrease in electrical resistance of RPAEC monolayer. These effects were partially eliminated in the presence of N,N'-bis (2-hydroxybenzyl) ethylenediamine-N,N'-diacetic acid, an iron chelator, and Y27632, a Rho-kinase inhibitor. Thus acute increases in labile iron in cultured pulmonary endothelium result in structural remodeling (and a proinflammatory phenotype) that occurs via post-transcriptional mechanisms regulated in a redox sensitive fashion.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Hierro/farmacología , Arteria Pulmonar/citología , Animales , Caveolina 1/metabolismo , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/citología , Glutatión/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Hierro/metabolismo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Methods Mol Biol ; 2454: 197-212, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33774811

RESUMEN

Studies have shown that human-induced pluripotent stem cells (iPSCs) derived cardiomyocytes (iCMCs) would provide a limitless source of cells for regenerative therapy and drug discoveries. Similar to embryonic stem cells, iPSCs have the capability to differentiate into mature functional iCMCs. The objective of our study is to develop an animal-free and viral-free approach by using a highly efficient transfection method that utilizes a critical combination of DNAs and mRNAs of pluripotent genes to generate iPSCs from adult human skin fibroblasts (SF). Subsequently differentiated them into functional cardiomyocytes. We obtained 4% of SFs into iPSCs at Passage 0, which shows significantly higher efficiency of reprogramming when compared to the use of either DNA alone or mRNAs alone. These iPSCs cultured under cardiac culture conditions are capable of differentiating into iCMCs. Furthermore, >88% of iCMCs are positive for either cardiac troponin T (TNNT2) or GATA binding protein 4 (GATA4). The iCMCs produced from SFs have been used in our laboratory to demonstrate their in vitro and in vivo functional potentials. In this study, we present step-by-step procedures for the generation of iPSCs from SFs and further differentiate them toward functional iCMCs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Adulto , Diferenciación Celular/genética , Células Cultivadas , Fibroblastos , Humanos , Miocitos Cardíacos , Piel
18.
Exp Biol Med (Maywood) ; 247(5): 433-445, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34648374

RESUMEN

The process of bone repair has always been a natural mystery. Although bones do repair themselves, supplemental treatment is required for the initiation of the self-regeneration process. Predominantly, surgical procedures are employed for bone regeneration. Recently, cell-based therapy for bone regeneration has proven to be more effective than traditional methods, as it eliminates the immune risk and painful surgeries. In clinical trials, various stem cells, especially mesenchymal stem cells, have shown to be more efficient for the treatment of several bone-related diseases, such as non-union fracture, osteogenesis imperfecta, osteosarcoma, and osteoporosis. Furthermore, the stem cells grown in a suitable three-dimensional scaffold support were found to be more efficient for osteogenesis. It has been shown that the three-dimensional bioscaffolds support and simulate an in vivo environment, which helps in differentiation of stem cells into bone cells. Bone regeneration in patients with bone disorders can be improved through modification of stem cells with several osteogenic factors or using stem cells as carriers for osteogenic factors. In this review, we focused on the various types of stem cells and scaffolds that are being used for bone regeneration. In addition, the molecular mechanisms of various transcription factors, signaling pathways that support bone regeneration and the senescence of the stem cells, which limits bone regeneration, have been discussed.


Asunto(s)
Osteogénesis , Andamios del Tejido , Regeneración Ósea , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre/metabolismo , Tecnología
19.
Cell Physiol Biochem ; 28(5): 911-22, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22178943

RESUMEN

Diabetic cardiomyopathy is associated with increased oxidative stress and inflammation. Mammalian 14-3-3 proteins are dimeric phosphoserine-binding proteins that participate in signal transduction and regulate several aspects of cellular biochemistry. The aim of the study presented here was to clarify the role of 14-3-3 protein in the mitogen activated protein kinase (MAPK) and nuclear factor-kB (NF-κB) signaling pathway after experimental diabetes by using transgenic mice with cardiac-specific expression of a dominant-negative 14-3-3 protein mutant (DN 14-3-3). Significant p-p38 MAPK activation in DN 14-3-3 mice compared to wild type mice (WT) after diabetes induction and with a corresponding up regulation of its downstream effectors, p-MAPK activated protein kinase 2 (MAPKAPK-2). Marked increases in cardiac hypertrophy, fibrosis and inflammation were observed with a corresponding up-regulation of atrial natriuretic peptide, osteopontin, connective tissue growth factor, tumor necrosis factor α, interleukin (IL)-1ß, IL-6 and cellular adhesion molecules. Moreover, reactive oxygen species, left ventricular expression of NADPH oxidase subunits, p22 phox, p67 phox, and Nox4, and lipid peroxidation levels were significantly increased in diabetic DN 14-3-3mice compared to diabetic WT mice. Furthermore, myocardial NF-κB activation, inhibitor of kappa B-α degradation and mRNA expression of proinflammatory cytokines were significantly increased in DN 14-3-3 mice compared to WT mice after diabetes induction. In conclusion, our data suggests that depletion of 14-3-3 protein induces cardiac oxidative stress, inflammation and remodeling after experimental diabetes induction mediated through p38 MAPK, MAPKAPK-2 and NF-κB signaling.


Asunto(s)
Diabetes Mellitus Experimental , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Transducción de Señal , Remodelación Ventricular , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Animales , Cardiomegalia/metabolismo , Moléculas de Adhesión Celular/metabolismo , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Regulación de la Expresión Génica , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Estreptozocina , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Cell Mol Med ; 14(9): 2235-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20716127

RESUMEN

To study the efficiency of maintaining the reduced tissue environment via pre-treatment with natural antioxidant resveratrol in stem cell therapy, we pre-treated male Sprague-Dawley rats with resveratrol (2.5 mg/kg/day gavaged for 2 weeks). After occlusion of the left anterior descending coronary artery (LAD), adult cardiac stem cells stably expressing EGFP were injected into the border zone of the myocardium. One week after the LAD occlusion, the cardiac reduced environment was confirmed in resveratrol-treated rat hearts by the enhanced expression of nuclear factor-E2-related factor-2 (Nrf2) and redox effector factor-1 (Ref-1). In concert, cardiac functional parameters (left ventricular ejection fraction and fractional shortening) were significantly improved. The improvement of cardiac function was accompanied by the enhanced stem cell survival and proliferation as demonstrated by the expression of cell proliferation marker Ki67 and differentiation of stem cells towards the regeneration of the myocardium as demonstrated by the enhanced expression of EGFP 28 days after LAD occlusion in the resveratrol-treated hearts. Our results demonstrate that resveratrol maintained a reduced tissue environment by overexpressing Nrf2 and Ref-1 in rats resulting in an enhancement of the cardiac regeneration of the adult cardiac stem cells as demonstrated by increased cell survival and differentiation leading to cardiac function.


Asunto(s)
Antioxidantes/farmacología , Infarto del Miocardio/terapia , Miocardio/patología , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos , Estilbenos/farmacología , Vino , Animales , Quimiocina CXCL12/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Pruebas de Función Cardíaca , Masculino , Infarto del Miocardio/fisiopatología , Ratas , Ratas Sprague-Dawley , Resveratrol , Trasplante de Células Madre , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA