Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Semin Cell Dev Biol ; 128: 145-153, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34219034

RESUMEN

Keratin (KRT), a natural fibrous structural protein, can be classified into two categories: "soft" cytosolic KRT that is primarily found in the epithelia tissues (e.g., skin, the inner lining of digestive tract) and "hard" KRT that is mainly found in the protective tissues (e.g., hair, horn). The latter is the predominant form of KRT widely used in biomedical research. The oxidized form of extracted KRT is exclusively denoted as keratose (KOS) while the reduced form of KRT is termed as kerateine (KRTN). KOS can be processed into various forms (e.g., hydrogel, films, fibers, and coatings) for different biomedical applications. KRT/KOS offers numerous advantages over other types of biomaterials, such as bioactivity, biocompatibility, degradability, immune/inflammatory privileges, mechanical resilience, chemical manipulability, and easy accessibility. As a result, KRT/KOS has attracted considerable attention and led to a large number of publications associated with this biomaterial over the past few decades; however, most (if not all) of the published review articles focus on KRT regarding its molecular structure, biochemical/biophysical properties, bioactivity, biocompatibility, drug/cell delivery, and in vivo transplantation, as well as its applications in biotechnical products and medical devices. Current progress that is directly associated with KOS applications in tissue regeneration and drug delivery appears an important topic that merits a commentary. To this end, the present review aims to summarize the current progress of KOS-associated biomedical applications, especially focusing on the in vitro and in vivo effects of KOS hydrogel on cultured cells and tissue regeneration following skin injury, skeletal muscle loss, peripheral nerve injury, and cardiac infarction.


Asunto(s)
Hidrogeles , Queratosis , Materiales Biocompatibles/análisis , Cabello/química , Humanos , Hidrogeles/análisis , Hidrogeles/química , Queratinas/análisis , Queratinas/química , Queratinas/farmacología
2.
Semin Cell Dev Biol ; 100: 52-61, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31623926

RESUMEN

Regeneration of cardiomyocytes, endothelial cells and vascular smooth muscle cells (three major lineages of cardiac tissues) following myocardial infarction is the critical step to recover the function of the damaged heart. Myeloid ecotropic viral integration site 1 (Meis1) was first discovered in leukemic mice in 1995 and its biological function has been extensively studied in leukemia, hematopoiesis, the embryonic pattering of body axis, eye development and various genetic diseases, such as restless leg syndrome. It was found that Meis1 is highly associated with Hox genes and their cofactors to exert its regulatory effects on multiple intracellular signaling pathways. Recently with the advent of bioinformatics, biochemical methods and advanced genetic engineering tools, new function of Meis1 has been found to be involved in the cell cycle regulation of cardiomyocytes and endothelial cells. For example, inhibition of Meis1 expression increases the proliferative capacity of neonatal mouse cardiomyocytes, whereas overexpression of Meis1 results in the reduction in the length of cardiomyocyte proliferative window. Interestingly, downregulation of one of the circular RNAs, which acts downstream of Meis1 in the cardiomyocytes, promotes angiogenesis and restores the myocardial blood supply, thus reinforcing better regeneration of the damaged heart. It appears that Meis1 may play double roles in modulating proliferation and regeneration of cardiomyocytes and endothelial cells post-myocardial infarction. In this review, we propose to summarize the major findings of Meis1 in modulating fetal development and adult abnormalities, especially focusing on the recent discoveries of Meis1 in controlling the fate of cardiomyocytes and endothelial cells.


Asunto(s)
Genes Homeobox , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Regeneración , Animales , Humanos
3.
Rev Cardiovasc Med ; 23(3): 97, 2022 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-35345264

RESUMEN

Toll-like receptors (TLRs) and interleukin-1 receptor (IL-1R) directly interact with intracellular interleukin receptor associated kinase (IRAK) family members to initialize innate immune and inflammatory responses following activation by pathogen-associated or host-derived elements. Although four IRAK family members [IRAK1, 2, 3 (i.e., IRAK-M), and 4] are involved in TLR and IL-1R signaling pathways, IL-1R > IRAK1 signaling appears to be the most studied pathway, with sufficient evidence to support its central role linking the innate immune response to the pathogenesis of various diseases, including cancers, metabolic disorders, and non-infectious immune disorders. However, IRAK1's involvement in cardiovascular diseases was only recently revealed and the detailed mechanism underling the pathogenesis of cardiovascular diseases, such as atherosclerosis, myocardial infarction, and heart failure (all non-infectious disorders), remains largely unknown with very limited publications to date. This review aims to summarize the overall roles of the IRAK family, especially IRAK1, in mediating the development of cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Quinasas Asociadas a Receptores de Interleucina-1 , Enfermedades Cardiovasculares/diagnóstico , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Receptores de Interleucina , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
5.
Microvasc Res ; 110: 48-55, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27998712

RESUMEN

BACKGROUND AND AIM: Although femoral artery ligation-induced ischemia is commonly used in C57BL/6 or Balb/c mice, direct comparisons between femoral artery/vein (FAV) versus femoral artery (FA) excisions have not been reported. The goal of the present study is to investigate the effects of FAV versus FA excisions on hindlimb models using adult CD-1 mice. METHODS: Two groups (n=10/group) of adult, mixed gender CD-1 mice were used to generate hindlimb ischemic models by excising either the FAV or FA. Laser Doppler Imaging was used to evaluate blood flow before surgery, immediately after surgery (Day 0), and then on Days 14 and 28. Toe necrosis was checked every 14days while skeletal muscle cellular remodeling and vascular networks were analyzed at the end of the experiment using pathohistological, Dil-vessel painting, and immunohistochemical approaches. RESULTS: During the 4-week period, no statistical differences were found between FAV and FA excision-induced ischemia in terms of reduction of limb blood flow, paw size, number of necrotic toes, or skeletal muscle cell sizes. However, significant increases in centrally-located nuclei cells, adipose cells, diameters of Dil-stained arterioles, and CD31+ capillary densities, but decreases in arteriole densities/lengths were observed in ischemic limbs of both FAV and FA groups compared to control limbs. CONCLUSION: We conclude that FAV and FA excision in CD-1 mice generate a comparable degree of hindlimb ischemia, suggesting that, as expected, FAV is no more severe than FA. These findings may provide important information for researchers when selecting ligation methods for their hindlimb models.


Asunto(s)
Capilares/fisiopatología , Arteria Femoral/cirugía , Vena Femoral/cirugía , Isquemia/fisiopatología , Músculo Esquelético/irrigación sanguínea , Adaptación Fisiológica , Animales , Velocidad del Flujo Sanguíneo , Capilares/metabolismo , Capilares/patología , Modelos Animales de Enfermedad , Electrocoagulación , Femenino , Arteria Femoral/fisiopatología , Vena Femoral/fisiopatología , Miembro Posterior , Isquemia/etiología , Isquemia/metabolismo , Isquemia/patología , Flujometría por Láser-Doppler , Masculino , Ratones , Músculo Esquelético/patología , Necrosis , Neovascularización Fisiológica , Imagen de Perfusión/métodos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Flujo Sanguíneo Regional , Factores de Tiempo
6.
Exp Cell Res ; 347(1): 143-152, 2016 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-27481582

RESUMEN

Electrospun microfiber meshes have been shown to support the proliferation and differentiation of many types of stem cells, but the phenotypic fate of c-kit(+) human cardiac stem cells (hCSCs) have not been explored. To this end, we utilized thin (~5µm) elastomeric meshes consisting of aligned 1.7µm diameter poly (ester-urethane urea) microfibers as substrates to examine their effect on hCSC viability, morphology, proliferation, and differentiation relative to cells cultured on tissue culture polystyrene (TCPS). The results showed that cells on microfiber meshes displayed an elongated morphology aligned in the direction of fiber orientation, lower proliferation rates, but increased expressions of genes and proteins majorly associated with cardiomyocyte phenotype. The early (NK2 homeobox 5, Nkx2.5) and late (cardiac troponin I, cTnI) cardiomyocyte genes were significantly increased on meshes (Nkx=2.5 56.2±13.0, cTnl=2.9±0.56,) over TCPS (Nkx2.5=4.2±0.9, cTnl=1.6±0.5, n=9, p<0.05 for both groups) after differentiation. In contrast, expressions of smooth muscle markers, Gata6 and myosin heavy chain (SM-MHC), were decreased on meshes. Immunocytochemical analysis with cardiac antibody exhibited the similar pattern of above cardiac differentiation. We conclude that aligned microfiber meshes are suitable for guiding cardiac differentiation of hCSCs and may facilitate stem cell-based therapies for treatment of cardiac diseases.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Miocardio/citología , Poliésteres/farmacología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/citología , Andamios del Tejido/química , Biomarcadores/metabolismo , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Células Madre/efectos de los fármacos , Células Madre/metabolismo
7.
Biochem Biophys Res Commun ; 473(4): 907-912, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27040769

RESUMEN

Previous studies indicated that the fast-healer strain of MRL/MpJ-Fas(lpr)/J (MRL) mice demonstrated superior regenerative capabilities for digit wound healing and/or regeneration compared with the non-healer strain of C57BL/6 (C57) mice. These reports, however, mainly focused on morphological observations and analysis of gene expression with little attention on the role of angiogenesis in the amputated digits. By taking advantage of Laser Doppler Imaging and histological analysis, we examined the potential role(s) of angiogenesis in facilitating tissue regrowth/regeneration by comparing two strains of mice (MRL versus C57). The three middle digits on the mouse's right foot (RF) were amputated at the middle level of phalanx 2 (P2) on postnatal day 2 (Day 0), while the left foot (LF) remained intact and served as a control. Laser Doppler images and digital photographs were taken of both feet before, immediately after surgery, and on Day 7, 14, 21, and 28 to evaluate blood flow and overall length of digit regrowth. All measurements from the amputated digits of the RF were divided by those of the control LF to obtain normalized ratios for statistical comparisons between groups. It was found that MRL mice demonstrated an approximately 220% increase in regrowth ratios over that of C57 mice from Day 21-28 (p < 0.01, n = 13), while blood-flow increased by about 25% on Day 21 (p < 0.01, n = 13) compared to that in C57 mice. Histological analysis of both control and amputated limbs indicated an approximately 70% increase in the number of vessels (both arterial and venous) in MRL mice over that of the C57 mice (p < 0.05, n = 3). We conclude that higher blood flow and angiogenesis may play an important role in facilitating the fast regrowth ratios of amputated digits in MRL mice compared to C57 mice.


Asunto(s)
Neovascularización Fisiológica , Dedos del Pie/fisiología , Cicatrización de Heridas , Animales , Pie/diagnóstico por imagen , Flujometría por Láser-Doppler , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Flujo Sanguíneo Regional , Dedos del Pie/irrigación sanguínea , Dedos del Pie/diagnóstico por imagen
8.
J Xray Sci Technol ; 24(4): 549-63, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-27163376

RESUMEN

BACKGROUND: The relatively high radiation dose from micro-CT is a cause for concern in preclinical research involving animal subjects. Interior region-of-interest (ROI) imaging was proposed for dose reduction, but has not been experimentally applied in micro-CT. OBJECTIVE: Our aim is to implement interior ROI imaging in a carbon nanotube (CNT) x-ray source based micro-CT, and present the ROI image quality and radiation dose reduction for interior cardiac micro-CT imaging of a mouse heart in situ. METHODS: An aperture collimator was mounted at the source-side to induce a small-sized cone beam (10 mm width) at the isocenter. Interior in situ micro-CT scans were conducted on a mouse carcass and several micro-CT phantoms. A GPU-accelerated hybrid iterative reconstruction algorithm was employed for volumetric image reconstruction. Radiation dose was measured for the same system operated at the interior and global micro-CT modes. RESULTS: Visual inspection demonstrated comparable image quality between two scan modes. Quantitative evaluation demonstrated high structural similarity index (up to 0.9614) with improved contrast-noise-ratio (CNR) on interior micro-CT mode. Interior micro-CT mode yielded significant reduction (up to 83.9%) for dose length product (DLP). CONCLUSIONS: This work demonstrates the applicability of using CNT x-ray source based interior micro-CT for preclinical imaging with significantly reduced radiation dose.


Asunto(s)
Corazón/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Nanotubos de Carbono/química , Microtomografía por Rayos X/métodos , Algoritmos , Animales , Ratones , Fantasmas de Imagen
10.
ACS Pharmacol Transl Sci ; 7(5): 1624-1636, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38751644

RESUMEN

In previous research, a synthetic α-carboxyl terminus 1 (αCT1) peptide derived from connexin 43 (Cx43) and its variant (αCT11) showed beneficial effects in an ex vivo ischemia-reperfusion (I/R) heart injury model in mouse. In an in vivo mouse model of cryo-induced ventricular injury, αCT1 released from adhesive cardiac patches reduced Cx43 remodeling and arrhythmias, as well as maintained cardiac conduction. Whether intravenous injection of αCT1 or αCT11 produces similar outcomes has not been investigated. Given the possibility of peptide degradation in plasma, this study utilized in vivo I/R cardiac injury and ex vivo blood plasma models to examine factors that may limit the therapeutic potential of peptide therapeutics in vivo. Following tail vein administration of αCT11 (100 µM) in blood, no effect on I/R infarct size was observed in adult rat hearts on day 1 (D1) and day 28 (D28) after injury (p > 0.05). There was also no difference in the echocardiographic ejection fraction (EF%) between the control and the αCT11 groups (p > 0.05). Surprisingly, αCT11 in blood plasma collected from these rats was undetectable within ∼10 min after tail vein injection. To investigate factors that may modulate αCT11 degradation in blood, αCT11 was directly added to blood plasma isolated from normal rats without I/R and peptide levels were measured under different experimental conditions. Consistent with in vivo observations, significant αCT11 degradation occurred in plasma within 10 min at 22 and 37 °C and was nearly undetectable by 30 min. These responses were reduced by the addition of protease/phosphatase (PTase/PPTase) inhibitors to the isolated plasma. Interestingly, no significant differences in αCT11 degradation in plasma were noted between male and female rats. We conclude that fast degradation of αCT11 is likely the reason that no beneficial effects were observed in the in vivo I/R model and inhibition or shielding from PTase/PPTase activity may be a strategy that will assist with the viability of peptide therapeutics.

11.
J Biol Chem ; 287(40): 33720-32, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22879597

RESUMEN

Intracoronary delivery of c-kit-positive human cardiac stem cells (hCSCs) is a promising approach to repair the infarcted heart, but it is severely limited by the poor survival of donor cells. Cobalt protoporphyrin (CoPP), a well known heme oxygenase 1 inducer, has been used to promote endogenous CO generation and protect against ischemia/reperfusion injury. Therefore, we determined whether preconditioning hCSCs with CoPP promotes CSC survival. c-kit-positive, lineage-negative hCSCs were isolated from human heart biopsies. Lactate dehydrogenase release assays demonstrated that preconditioning CSCs with CoPP markedly enhanced cell survival after oxidative stress induced by H(2)O(2), concomitant with up-regulation of heme oxygenase 1, COX-2, and anti-apoptotic proteins (BCL2, BCL2-A1, and MCL-1) and increased phosphorylation of NRF2. Apoptotic cytometric assays showed that pretreatment of CSCs with CoPP enhanced the cells' resistance to apoptosis induced by oxidative stress. Conversely, knocking down HO-1, COX-2, or NRF2 by shRNA gene silencing abrogated the cytoprotective effects of CoPP. Further, preconditioning CSCs with CoPP led to a global increase in release of cytokines, such as EGF, FGFs, colony-stimulating factors, and chemokine ligand. Conditioned medium from cells pretreated with CoPP conferred naive CSCs remarkable resistance to apoptosis, demonstrating that cytokines released by preconditioned cells play a key role in the anti-apoptotic effects of CoPP. Preconditioning CSCs with CoPP also induced an increase in the phosphorylation of Erk1/2, which are known to modulate multiple pro-survival genes. These results potentially provide a simple and effective strategy to enhance survival of CSCs after transplantation and, therefore, their efficacy in repairing infarcted myocardium.


Asunto(s)
Apoptosis , Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Cardiopatías/tratamiento farmacológico , Hemo-Oxigenasa 1/química , Miocardio/citología , Factor 2 Relacionado con NF-E2/metabolismo , Protoporfirinas/farmacología , Células Madre/citología , Humanos , L-Lactato Deshidrogenasa/metabolismo , Lentivirus/metabolismo , Modelos Genéticos , Isquemia Miocárdica/patología , Estrés Oxidativo , ARN Interferente Pequeño/metabolismo
12.
Stem Cell Rev Rep ; 19(7): 2341-2360, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37392292

RESUMEN

Peripheral artery disease (PAD) is a common vascular disorder in the extremity of limbs with limited clinical treatments. Stem cells hold great promise for the treatment of PAD, but their therapeutic efficiency is limited due to multiple factors, such as poor engraftment and non-optimal selection of cell type. To date, stem cells from a variety of tissue sources have been tested, but little information is available regarding vascular smooth muscle cells (VSMCs) for PAD therapy. The present study examines the effects of keratose (KOS) hydrogels on c-kit+/CD31- cardiac vascular smooth muscle progenitor cell (cVSMPC) differentiation and the therapeutic potential of the resultant VSMCs in a mouse hindlimb ischemic model of PAD. The results demonstrated that KOS but not collagen hydrogel was able to drive the majority of cVSMPCs into functional VSMCs in a defined Knockout serum replacement (SR) medium in the absence of differentiation inducers. This effect could be inhibited by TGF-ß1 antagonists. Further, KOS hydrogel increased expression of TGF-ß1-associated proteins and modulated the level of free TGF-ß1 during differentiation. Finally, transplantation of KOS-driven VSMCs significantly increased blood flow and vascular densities of ischemic hindlimbs. These findings indicate that TGF-ß1 signaling is involved in KOS hydrogel-preferred VSMC differentiation and that enhanced blood flow are likely resulted from angiogenesis and/or arteriogenesis induced by transplanted VSMCs.

13.
Curr Vasc Pharmacol ; 20(1): 29-36, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34387163

RESUMEN

Trimethylamine N-oxide (TMAO) is a gut microbiota metabolite derived from trimethylamine- containing nutrient precursors such as choline, L-carnitine, and betaine, which are rich in many vegetables, fruits, nuts, dairy products, and meats. An increasing number of clinical studies have demonstrated a strong relationship between elevated plasma TMAO levels and adverse cardiovascular events. It is commonly agreed that TMAO acts as an independent risk factor and a prognostic index for patients with cardiovascular disease. Although most animal (mainly rodent) data support the clinical findings, the mechanisms by which TMAO modulates the cardiovascular system are still not well understood. In this context, we provide an overview of the potential mechanisms underlying TMAO-induced cardiovascular diseases at the cellular and molecular levels, with a focus on atherosclerosis. We also address the direct effects of TMAO on cardiomyocytes (a new and under-researched area) and finally propose TMAO as a potential biomarker and/or therapeutic target for diagnosis and treatment of patients with cardiovascular disease.


Asunto(s)
Aterosclerosis , Cardiomiopatías , Enfermedades Cardiovasculares , Animales , Aterosclerosis/diagnóstico , Betaína/efectos adversos , Enfermedades Cardiovasculares/diagnóstico , Humanos , Metilaminas/metabolismo
14.
Eur J Pharmacol ; 932: 175192, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-35981605

RESUMEN

Current medicinal treatments for diseases comprise largely of two categories: small molecular (chemical) (e.g., aspirin) and larger molecular (peptides/proteins, e.g., insulin) drugs. Whilst both types of therapeutics can effectively treat different diseases, ranging from well-understood (in view of pathogenesis and treatment) examples (e.g., flu), to less-understood chronic diseases (e.g., diabetes), classical small molecule drugs often possess significant side-effects (a major cause of drug withdrawal from market) due to their low- or non-specific targeting. By contrast, therapeutic peptides, which comprise short sequences from naturally occurring peptides/proteins, commonly demonstrate high target specificity, well-characterized modes-of-action, and low or non-toxicity in vivo. Unfortunately, due to their small size, linear permutation, and lack of tertiary structure, peptidic drugs are easily subject to rapid degradation or loss in vivo through chemical and physical routines, thus resulting in a short half-life and reduced therapeutic efficacy, a major drawback that can reduce therapeutic efficiency. However, recent studies demonstrate that the short half-life of peptidic drugs can be significantly extended by various means, including use of enantiomeric or non-natural amino acids (AAs) (e.g., L-AAs replacement with D-AAs), chemical conjugation [e.g., with polyethylene glycol], and encapsulation (e.g., in exosomes). In this context, we provide an overview of the major in vivo degradation forms of small therapeutic peptides in the plasma and anti-degradation strategies. We also update on the progress of small peptide therapeutics that are either currently in clinical trials or are being successfully used in clinical therapies for patients with non-infectious diseases, such as diabetes, multiple sclerosis, and cancer.


Asunto(s)
Diabetes Mellitus , Insulinas , Enfermedades no Transmisibles , Aminoácidos , Aspirina , Humanos , Péptidos/química , Péptidos/farmacología , Péptidos/uso terapéutico , Polietilenglicoles , Proteínas
15.
EXCLI J ; 20: 126-141, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33564282

RESUMEN

Coronary artery disease (CAD) and atrial fibrillation (AF) share common risk factors, such as hypertension and diabetes. The patients with CAD often suffer concomitantly AF, but how two diseases interact with each other at cellular and molecular levels remain largely unknown. The present study aims to dissect the common differentially expressed genes (DEGs) that are concurrently associated with CAD and AF. Two datasets [GSE71226 for CAD) and GSE31821 for AF] were analyzed with GEO2R and Venn Diagram to identify the DEGs. Signaling pathways, gene enrichments, and protein-protein interactions (PPI) of the identified common DEGs were further analyzed with Kyoto Encyclopedia of Gene and Genome (KEGG), Database for Annotation, Visualization and Integrated Discovery (DAVID), and Search Toll for the Retrieval of Interacting Genes (STRING). 565 up- and 1367 down-regulated genes in GSE71226 and 293 up- and 68 down-regulated genes in GSE31821 were identified. Among those, 21 common DEGs were discovered from both datasets, which lead to the findings of 4 CAD and 21 AF pathways, 3 significant gene enrichments (intracellular cytoplasm, protein binding, and vascular labyrinthine layer), and 3 key proteins (membrane metallo-endopeptidase (MME), transferrin receptor 1 (TfR1), and Lysosome-associated membrane glycoprotein 1 (LAMP1)). Together, these data implied that these three proteins may play a central role in development of both CAD and AF.

16.
Circ Res ; 103(7): e81-95, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18776039

RESUMEN

Cardiac I Kr is a critical repolarizing current in the heart and a target for inherited and acquired long-QT syndrome (LQTS). Biochemical and functional studies have demonstrated that I Kr channels are heteromers composed of both hERG 1a and 1b subunits, yet our current understanding of I Kr functional properties derives primarily from studies of homooligomers of the original hERG 1a isolate. Here, we examine currents produced by hERG 1a and 1a/1b channels expressed in HEK-293 cells at near-physiological temperatures. We find that heteromeric hERG 1a/1b currents are much larger than hERG 1a currents and conduct 80% more charge during an action potential. This surprising difference corresponds to a 2-fold increase in the apparent rates of activation and recovery from inactivation, thus reducing rectification and facilitating current rebound during repolarization. Kinetic modeling shows these gating differences account quantitatively for the differences in current amplitude between the 2 channel types. Drug sensitivity was also different. Compared to homomeric 1a channels, heteromeric 1a/1b channels were inhibited by E-4031 with a slower time course and a corresponding 4-fold shift in the IC50. The importance of hERG 1b in vivo is supported by the identification of a 1b-specific A8V missense mutation in 1/269 unrelated genotype-negative LQTS patients that was absent in 400 control alleles. Mutant 1bA8V expressed alone or with hERG 1a in HEK-293 cells dramatically reduced 1b protein levels. Thus, mutations specifically disrupting hERG 1b function are expected to reduce cardiac I Kr and enhance drug sensitivity, and represent a potential mechanism underlying inherited or acquired LQTS.


Asunto(s)
Sustitución de Aminoácidos , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Mutación Missense , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Antiarrítmicos/farmacología , Línea Celular , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/genética , Cinética , Modelos Biológicos , Piperidinas , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Piridinas
17.
Biofabrication ; 12(3): 035026, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32434163

RESUMEN

3D bioprinting is an emerging biofabrication process for the production of adherent cell-based products, including engineered tissues and foods. While process innovations are rapidly occurring in the area of process monitoring, which can improve fundamental understanding of process-structure-property relations as well as product quality by closed-loop control techniques, in-line sensing of the bioink composition remains a challenge. Here, we report that hollow multifunctional fibers enable in-line impedimetric sensing of bioink composition and exhibit selectivity for real-time classification of cell type, viability, and state of differentiation during bioprinting. Continuous monitoring of the fiber impedance magnitude and phase angle response from 102 to 106 Hz during microextrusion 3D bioprinting enabled compositional and quality analysis of alginate bioinks that contained fibroblasts, neurons, or mouse embryonic stem cells (mESCs). Fiber impedimetric responses associated with the bioinks that contained differentiated mESCs were consistent with differentiation marker expression characterized by immunocytochemistry. 3D bioprinting through hollow multifunctional fiber impedimetric sensors enabled classification of stem cells as stable or randomly differentiated populations. This work reports an advance in monitoring 3D bioprinting processes in terms of in-line sensor-based bioink compositional analysis using fiber technology and provides a non-invasive sensing platform for achieving future quality-controlled bioprinted tissues and injectable stem-cell therapies.


Asunto(s)
Bioimpresión , Técnicas Biosensibles , Impedancia Eléctrica , Impresión Tridimensional , Animales , Diferenciación Celular , Supervivencia Celular , Electrodos , Tinta , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Células PC12 , Ratas , Transducción de Señal , Células Madre/citología
18.
Circ Res ; 97(6): 566-73, 2005 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-16100050

RESUMEN

The mechanisms underlying the blunted contractile response to beta-adrenergic receptor (beta-AR) stimulation in heart failure (HF) are incompletely understood, especially with regard to beta-AR subtype-specific regulation of L-type Ca2+ channels. We evaluated the impact of HF induced by pacing tachycardia on beta-AR regulation of L-type Ca2+ channels in a canine model. To evaluate changes in the relative subcellular distribution of beta-AR subtypes, left ventricular membranes enriched in surface sarcolemma and T-tubular sarcolemma were prepared. Radioligand binding using [(125)I]cyanopindolol revealed that HF resulted in a comparable decrease in the density of beta1-ARs in both surface and T-tubule sarcolemma (55+/-4%, n=7, P<0.001; and 45+/-10%, n=7, P<0.01, respectively), but no significant change in beta2-AR density was observed. Whole-cell patch clamp studies demonstrated a markedly blunted increase in I(Ca,L) in response to saturating concentrations of the nonselective beta-AR agonist isoproterenol (0.1 micromol/L) in failing myocytes compared with control (129+/-20%, n=11, versus 332+/-35%, n=7; P<0.001). Experiments testing beta1-AR- and beta2-AR-selective stimulation showed that the major component of the blunted response to nonselective beta-AR stimulation in HF was caused by beta2-AR activation, resulting in a pertussis toxin-sensitive, Gi-mediated inhibition of the beta1-AR-induced increase in I(Ca,L). In conclusion, canine HF results in the following: (1) a uniform reduction in beta1-AR density in surface and T-tubule membrane fractions without a change in beta2-AR density; and (2) the emergence of distinct Gi-coupling to beta2-ARs resulting in accentuated antagonism of beta1-AR-mediated stimulation of I(Ca,L). These results have implications for optimizing the use of beta-AR drugs in HF.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Insuficiencia Cardíaca/fisiopatología , Receptores Adrenérgicos beta 1/fisiología , Receptores Adrenérgicos beta 2/fisiología , Transducción de Señal/fisiología , Albuterol/farmacología , Animales , Perros , Isoproterenol/farmacología , Toxina del Pertussis/farmacología , Propanolaminas/farmacología , Receptores Adrenérgicos beta 1/análisis , Receptores Adrenérgicos beta 2/análisis , Sarcolema/química
19.
J Biomater Sci Polym Ed ; 28(13): 1245-1255, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28391767

RESUMEN

Alginate-based microencapsulation of cells has made a significant impact on the fields of regenerative medicine and tissue engineering mainly because of its ability to provide immunoisolation for the encapsulated material. This characteristic has allowed for the successful transplantation of non-autologous cells in several clinical trials for life threatening conditions, such as diabetes, myocardial infarction, and neurodegenerative disorders. Methods for alginate hydrogel microencapsulation have been well developed for various types of cells and can generate microcapsules of different diameters, degradation time, and composition. It appears the most prominent and successful method in clinical applications is the coaxial electrospray method, which can be used to generate both homogenous and non-homogeneous microcapsules with uniform size on the order of 100 µm. The present review aims to discuss why alginate hydrogel is an ideal biomaterial for the encapsulation of cells, how alginate-based microcapsules are generated, and methods of modifying the microcapsules for specific clinical treatments. This review will also discuss clinical applications that have utilized alginate-based microencapsulation in the treatment of diabetes, ischemic heart disease, and neurodegenerative diseases.


Asunto(s)
Alginatos , Electricidad , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Alginatos/química , Animales , Cápsulas , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos
20.
Stem Cells Dev ; 26(12): 888-900, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28351290

RESUMEN

Stem cell-based therapies have demonstrated great potential for the treatment of cardiac diseases, for example, myocardial infarction; however, low cell viability, low retention/engraftment, and uncontrollable in vivo differentiation after transplantation are still major limitations, which lead to low therapeutic efficiency. Biomaterials provide a promising solution to overcome these issues due to their biocompatibility, biodegradability, low/nonimmunogenicity, and low/noncytotoxicity. The present study aimed to investigate the impacts of keratose (KOS) hydrogel biomaterial on cellular viability, proliferation, and differentiation of c-kit+ human cardiac stem cells (hCSCs). Briefly, hCSCs were cultured on both KOS hydrogel-coated dishes and regular tissue culture dishes (Blank control). Cell viability, stemness, proliferation, cellular morphology, and cardiac lineage differentiation were compared between KOS hydrogel and the Blank control at different time points. We found that KOS hydrogel is effective in maintaining hCSCs without any observable toxic effects, although cell size and proliferation rate appeared smaller on the KOS hydrogel compared to the Blank control. To our surprise, KOS hydrogel significantly promoted vascular smooth muscle cell (VSMC) differentiation (∼72%), while on the Blank control dishes, most of the hCSCs (∼78%) became cardiomyocytes. Furthermore, we also observed "endothelial cell tube-like" microstructures formed by differentiated VSMCs only on KOS hydrogel, suggesting a potential capability of the hCSC-derived VSMCs for in vitro angiogenesis. To the best of our knowledge, this is the first report to discover the preferred differentiation of hCSCs toward VSMCs on KOS hydrogel. The underlying mechanism remains unknown. This innovative methodology may offer a new approach to generate a robust number of VSMCs simply by culturing hCSCs on KOS hydrogel, and the resulting VSMCs may be used in animal studies and clinical trials in combination with an injectable KOS hydrogel to treat cardiovascular diseases.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Hidrogeles/química , Miocardio , Miocitos Cardíacos , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Células Madre , Supervivencia Celular , Células Cultivadas , Humanos , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre/citología , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA