Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 326(1): G38-G52, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37933466

RESUMEN

In esophageal epithelial cells in eosinophilic esophagitis (EoE), Th2 cytokines (IL-4, IL-13) signal through IL-4Rα, activating JAK to increase eotaxin-3 secretion, which draws eosinophils into the mucosa. We explored whether Th2 cytokines also might stimulate eotaxin-3 secretion and increase tension in esophageal smooth muscle (ESM), which might impair esophageal distensibility, and whether those events could be blocked by proton pump inhibitors (PPIs) or agents that disrupt IL-4Rα signaling. We established human ESM cell cultures from organ donors, characterizing Th2 cytokine receptor and P-type ATPase expression by qPCR. We measured Th2 cytokine-stimulated eotaxin-3 secretion by enzyme-linked immunosorbent assay (ELISA) and ESM cell tension by gel contraction assay, before and after treatment with omeprazole, ruxolitinib (JAK inhibitor), or IL-4Rα blocking antibody. CPI-17 (inhibitor of a muscle-relaxing enzyme) effects were studied with CPI-17 knockdown by siRNA or CPI-17 phospho(T38A)-mutant overexpression. ESM cells expressed IL-4Rα and IL-13Rα1 but only minimal H+-K+-ATPase mRNA. Th2 cytokines increased ESM eotaxin-3 secretion and tension, effects blocked by ruxolitinib and IL-4Rα blocking antibody but not consistently blocked by omeprazole. IL-13 increased ESM tension by increasing CPI-17 expression and phosphorylation, effects blocked by CPI-17 knockdown. Blocking IL-4Rα decreased IL-13-stimulated eotaxin-3 secretion, CPI-17 expression, and tension in ESM. Th2 cytokines increase ESM eotaxin-3 secretion and tension via IL-4Rα signaling that activates CPI-17. Omeprazole does not reliably inhibit this process, but IL-4Rα blocking antibody does. This suggests that ESM eosinophilia and impaired esophageal distensibility might persist despite elimination of mucosal eosinophils by PPIs, and IL-4Rα blocking agents might be especially useful in this circumstance.NEW & NOTEWORTHY We have found that Th2 cytokines increase eotaxin-3 secretion and tension in esophageal smooth muscle (ESM) cells via IL-4Rα signaling. Unlike esophageal epithelial cells, ESM cells do not express H+-K+-ATPase, and omeprazole does not inhibit their cytokine-stimulated eotaxin-3 secretion or tension. An IL-4Rα blocking antibody reduces both eotaxin-3 secretion and tension induced by Th2 cytokines in ESM cells, suggesting that an agent such as dupilumab might be preferred for patients with EoE with esophageal muscle involvement.


Asunto(s)
Esofagitis Eosinofílica , Interleucina-13 , Humanos , Adenosina Trifosfatasas , Quimiocina CCL26 , Citocinas/metabolismo , Esofagitis Eosinofílica/metabolismo , Interleucina-13/farmacología , Músculo Liso/metabolismo , Omeprazol , Inhibidores de la Bomba de Protones/farmacología , Células Th2
2.
J Math Biol ; 88(2): 14, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38180543

RESUMEN

This study presents a new framework for obtaining personalized optimal treatment strategies targeting aberrant signaling pathways in esophageal cancer, such as the epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF) signaling pathways. A new pharmacokinetic model is developed taking into account specific heterogeneities of these signaling mechanisms. The optimal therapies are designed to be obtained using a three step process. First, a finite-dimensional constrained optimization problem is solved to obtain the parameters of the pharmacokinetic model, using discrete patient data measurements. Next, a sensitivity analysis is carried out to determine which of the parameters are sensitive to the evolution of the variants of EGF receptors and VEGF receptors. Finally, a second optimal control problem is solved based on the sensitivity analysis results, using a modified pharmacokinetic model that incorporates two representative drugs Trastuzumab and Bevacizumab, targeting EGF and VEGF, respectively. Numerical results with the combination of the two drugs demonstrate the efficiency of the proposed framework.


Asunto(s)
Factor de Crecimiento Epidérmico , Neoplasias Esofágicas , Humanos , Factor A de Crecimiento Endotelial Vascular , Transducción de Señal , Neoplasias Esofágicas/tratamiento farmacológico
3.
Am J Physiol Gastrointest Liver Physiol ; 322(6): G598-G614, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35380457

RESUMEN

The pathogenesis of subsquamous intestinal metaplasia (SSIM), in which glands of Barrett's esophagus (BE) are buried under esophageal squamous epithelium, is unknown. In a rat model of reflux esophagitis, we found that columnar-lined esophagus developed via a wound-healing process involving epithelial-mesenchymal plasticity (EMP) that buried glands under ulcerated squamous epithelium. To explore a role for reflux-induced EMP in BE, we established and characterized human Barrett's organoids and sought evidence of EMP after treatment with acidic bile salts (AB). We optimized media to grow human BE organoids from immortalized human Barrett's cells and from BE biopsies from seven patients, and we characterized histological, morphological, and molecular features of organoid development. Features and markers of EMP were explored following organoid exposure to AB, with and without a collagen I (COL1) matrix to simulate a wound-healing environment. All media successfully initiated organoid growth, but advanced DMEM/F12 (aDMEM) was best at sustaining organoid viability. Using aDMEM, organoids comprising nongoblet and goblet columnar cells that expressed gastric and intestinal cell markers were generated from BE biopsies of all seven patients. After AB treatment, early-stage Barrett's organoids exhibited EMP with loss of membranous E-cadherin and increased protrusive cell migration, events significantly enhanced by COL1. Using human BE biopsies, we have established Barrett's organoids that recapitulate key histological and molecular features of BE to serve as high-fidelity BE models. Our findings suggest that reflux can induce EMP in human BE, potentially enabling Barrett's cells to migrate under adjacent squamous epithelium to form SSIM.NEW & NOTEWORTHY Using Barrett's esophagus (BE) biopsies, we established organoids recapitulating key BE features. During early stages of organoid development, a GERD-like wound environment-induced features of epithelial-mesenchymal plasticity (EMP) in Barrett's progenitor cells, suggesting that reflux-induced EMP can enable Barrett's cells to migrate underneath squamous epithelium to form subsquamous intestinal metaplasia, a condition that may underlie Barrett's cancers that escape detection by endoscopic surveillance, and recurrences of Barrett's metaplasia following endoscopic eradication therapy.


Asunto(s)
Esófago de Barrett , Carcinoma de Células Escamosas , Neoplasias Esofágicas , Esofagitis Péptica , Reflujo Gastroesofágico , Animales , Esófago de Barrett/patología , Ácidos y Sales Biliares/farmacología , Carcinoma de Células Escamosas/complicaciones , Neoplasias Esofágicas/complicaciones , Neoplasias Esofágicas/patología , Reflujo Gastroesofágico/complicaciones , Humanos , Metaplasia , Organoides/patología , Ratas
4.
Gastroenterology ; 160(6): 2072-2088.e6, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33581123

RESUMEN

BACKGROUND & AIMS: In upper airway cells, T helper 2 cytokines that signal through interleukin-4 (IL-4) receptor-α have been shown to stimulate eotaxin-3 secretion via a nongastric proton pump (ngH+,K+ATPase). To seek novel targets for eosinophilic esophagitis (EoE) treatments, we evaluated ngH+,K+ATPase expression in EoE squamous cells, and explored molecular pathways involved in eotaxin-3 secretion by IL-4 receptor-α signaling. METHODS: ngH+,K+ATPase expression in EoE cells was evaluated by quantitative real-time polymerase chain reaction and Western blotting. IL-4-stimulated eotaxin-3 secretion was measured by enzyme-linked immunosorbent assay after treatment with omeprazole, SCH 28080 (potassium-competitive acid blocker), ethylene glycol-bis(ß-aminoethyl)-N,N,N',N'-tetraacetoxymethyl ester (calcium chelator), 2-aminoethoxydiphenyl borate (inhibitor of endoplasmic reticulum calcium release), verapamil, and diltiazem (L-type calcium channel inhibitors). Intracellular calcium transients were measured by Fluo-4 fluorescence. Key experiments were confirmed in EoE primary cells and in RNA sequencing datasets from mucosal biopsies of patients with EoE and controls. RESULTS: EoE cells expressed ngH+,K+ATPase messenger RNA and protein. Omeprazole and SCH 28080 decreased IL-4-stimulated eotaxin-3 secretion. IL-4 increased intracellular calcium transients, and IL-4-stimulated eotaxin-3 secretion was blocked by ethylene glycol-bis(ß-aminoethyl)-N,N,N',N'-tetraacetoxymethyl ester, 2-aminoethoxydiphenyl borate, verapamil, and diltiazem. The combination of omeprazole and verapamil suppressed IL-4-stimulated eotaxin-3 secretion more than either agent alone. EoE biopsies expressed higher ngH+,K+ATPase and exhibited more calcium signaling than controls. CONCLUSIONS: EoE cells express a nongastric proton pump that mediates T helper 2 cytokine-stimulated eotaxin-3 secretion. IL-4 induces calcium release from the endoplasmic reticulum and calcium entry via L-type calcium channels, increasing intracellular calcium that contributes to eotaxin-3 secretion by EoE cells. L-type calcium channel inhibitors block T helper 2 cytokine-stimulated eotaxin-3 secretion, suggesting a potential role for these agents in EoE treatment.


Asunto(s)
Quimiocina CCL26/metabolismo , Esofagitis Eosinofílica/metabolismo , Esofagitis Eosinofílica/patología , Células Epiteliales/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Transporte Biológico/efectos de los fármacos , Compuestos de Boro/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Línea Celular , Diltiazem/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Mucosa Esofágica/metabolismo , Mucosa Esofágica/patología , Famotidina/farmacología , Femenino , Antagonistas de los Receptores H2 de la Histamina/farmacología , Humanos , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Masculino , Omeprazol/farmacología , Cultivo Primario de Células , Inhibidores de la Bomba de Protones/farmacología , Bombas de Protones/efectos de los fármacos , Bombas de Protones/metabolismo , ARN Mensajero/metabolismo , Ranitidina/farmacología , Transducción de Señal/efectos de los fármacos , Células Th2/metabolismo , Verapamilo/farmacología
5.
J Math Biol ; 84(4): 23, 2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35212794

RESUMEN

In this paper, a new framework for obtaining personalized optimal treatment strategies in colon cancer-induced angiogenesis is presented. The dynamics of colon cancer is given by a Itó stochastic process, which helps in modeling the randomness present in the system. The stochastic dynamics is then represented by the Fokker-Planck (FP) partial differential equation that governs the evolution of the associated probability density function. The optimal therapies are obtained using a three step procedure. First, a finite dimensional FP-constrained optimization problem is formulated that takes input individual noisy patient data, and is solved to obtain the unknown parameters corresponding to the individual tumor characteristics. Next, a sensitivity analysis of the optimal parameter set is used to determine the parameters to be controlled, thus, helping in assessing the types of treatment therapies. Finally, a feedback FP control problem is solved to determine the optimal combination therapies. Numerical results with the combination drug, comprising of Bevacizumab and Capecitabine, demonstrate the efficiency of the proposed framework.


Asunto(s)
Neoplasias del Colon , Neoplasias del Colon/tratamiento farmacológico , Retroalimentación , Humanos , Procesos Estocásticos
6.
Int J Mol Sci ; 23(3)2022 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-35163685

RESUMEN

Targeting dysregulated Ca2+ signaling in cancer cells is an emerging chemotherapy approach. We previously reported that store-operated Ca2+ entry (SOCE) blockers, such as RP4010, are promising antitumor drugs for esophageal cancer. As a tyrosine kinase inhibitor (TKI), afatinib received FDA approval to be used in targeted therapy for patients with EGFR mutation-positive cancers. While preclinical studies and clinical trials have shown that afatinib has benefits for esophageal cancer patients, it is not known whether a combination of afatinib and RP4010 could achieve better anticancer effects. Since TKI can alter intracellular Ca2+ dynamics through EGFR/phospholipase C-γ pathway, in this study, we evaluated the inhibitory effect of afatinib and RP4010 on intracellular Ca2+ oscillations in KYSE-150, a human esophageal squamous cell carcinoma cell line, using both experimental and mathematical simulations. Our mathematical simulation of Ca2+ oscillations could fit well with experimental data responding to afatinib or RP4010, both separately or in combination. Guided by simulation, we were able to identify a proper ratio of afatinib and RP4010 for combined treatment, and such a combination presented synergistic anticancer-effect evidence by experimental measurement of intracellular Ca2+ and cell proliferation. This intracellular Ca2+ dynamic-based mathematical simulation approach could be useful for a rapid and cost-effective evaluation of combined targeting therapy drugs.


Asunto(s)
Afatinib/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Calcio/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Modelos Biológicos , Compuestos Orgánicos/uso terapéutico , Afatinib/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Neoplasias Esofágicas/patología , Humanos , Compuestos Orgánicos/farmacología
7.
Am J Physiol Gastrointest Liver Physiol ; 320(3): G319-G327, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33355505

RESUMEN

Mast cells and eosinophils are the key effector cells of allergic disorders. Although most studies on eosinophilic esophagitis (EoE), an allergic disorder of the esophagus, have focused on the role of eosinophils, recent studies suggest a major role for mast cells in causing the clinical manifestations of this disease. Cellular and animal studies have demonstrated that mast cells can cause esophageal muscle cells to proliferate and differentiate into a more contractile phenotype, and that mediators released by degranulating mast cells such as tryptase and histamine can activate smooth muscle contraction pathways. Thus, activated mast cells in the esophageal muscularis propria might cause esophageal motility abnormalities, including the failure of lower esophageal sphincter relaxation typical of achalasia. In addition, mast cells have been implicated in the pathogenesis of a number of neurodegenerative disorders of the central nervous system such as Alzheimer's and Parkinson's diseases, because degranulating mast cells release proinflammatory and cytotoxic mediators capable of damaging neurons. Such mast cell degranulation in the myenteric plexus of the esophagus could cause the loss of enteric neurons that characterizes achalasia. In this report, we review the molecular mechanisms of esophageal smooth muscle contraction, and how mast cells products might affect that muscle and cause neurodegeneration in the esophagus. Based on these data, we present our novel, conceptual model for an allergy-induced form of achalasia mediated by mast cell activation in the esophageal muscularis propria.


Asunto(s)
Esofagitis Eosinofílica/patología , Acalasia del Esófago/patología , Mastocitos/fisiología , Esófago/anatomía & histología , Esófago/inervación , Humanos , Músculo Liso/anatomía & histología , Músculo Liso/inervación
8.
Chembiochem ; 22(22): 3164-3168, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34506664

RESUMEN

The rational design of materials with cell-selective membrane activity is an effective strategy for the development of targeted molecular imaging and therapy. Here we report a new class of cationic multidomain peptides (MDPs) that can undergo enzyme-mediated molecular transformation followed by supramolecular assembly to form nanofibers in which cationic clusters are presented on a rigid ß-sheet backbone. This structural transformation, which is induced by cells overexpressing the specific enzymes, led to a shift in the membrane perturbation potential of the MDPs, and consequently enhanced cell uptake and drug delivery efficacy. We envision the directed self-assembly based on modularly designed MDPs as a highly promising approach to generate dynamic supramolecular nanomaterials with emerging membrane activity for a range of disease targeted molecular imaging and therapy applications.


Asunto(s)
Membrana Celular/metabolismo , Péptidos/metabolismo , Membrana Celular/química , Células HeLa , Humanos , Iones/síntesis química , Iones/química , Iones/metabolismo , Sustancias Macromoleculares/síntesis química , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Estructura Molecular , Tamaño de la Partícula , Péptidos/síntesis química , Péptidos/química
9.
Adv Exp Med Biol ; 1349: 303-333, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35138620

RESUMEN

Calcium (Ca2+) is a critical regulator of cardiovascular function. The Ca2+ channels, pumps, and exchangers contributing to cytosolic Ca2+ signals governing cardiac contraction and vascular tone are well known. In addition to these Ca2+ components, store-operated calcium entry (SOCE) is a ubiquitous mechanism recently recognized underlying cardiovascular function maintenance and disease development and progression. With this review article, we hope to highlight the accumulated knowledge about the SOCE machinery and its potential contribution to cardiac and vascular function and its roles in cardiovascular pathogenesis and pathology.


Asunto(s)
Calcio , Sistema Cardiovascular , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Sistema Cardiovascular/metabolismo , Molécula de Interacción Estromal 1/metabolismo
10.
Small ; 16(38): e2002780, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32812362

RESUMEN

Many new technologies, such as cancer microenvironment-induced nanoparticle targeting and multivalent ligand approach for cell surface receptors, are developed for active targeting in cancer therapy. While the principle of each technology is well illustrated, most systems suffer from low targeting specificity and sensitivity. To fill the gap, this work demonstrates a successful attempt to combine both technologies to simultaneously improve cancer cell targeting sensitivity and specificity. Specifically, the main component is a targeting ligand conjugated self-assembling monomer precursor (SAM-P), which, at the tumor site, undergoes tumor-triggered cleavage to release the active form of self-assembling monomer capable of forming supramolecular nanostructures. Biophysical characterization confirms the chemical and physical transformation of SAM-P from unimers or oligomers with low ligand valency to supramolecular assemblies with high ligand valency under a tumor-mimicking reductive microenvironment. The in vitro fluorescence assay shows the importance of supramolecular morphology in mediating ligand-receptor interactions and targeting sensitivity. Enhanced targeting specificity and sensitivity can be achieved via tumor-triggered supramolecular assembly and induces multivalent ligand presentation toward cell surface receptors, respectively. The results support this combined tumor microenvironment-induced cell targeting and multivalent ligand display approach, and have great potential for use as cell-specific molecular imaging and therapeutic agents with high sensitivity and specificity.


Asunto(s)
Nanofibras , Nanopartículas , Neoplasias , Humanos , Ligandos , Péptidos , Microambiente Tumoral
11.
FASEB J ; 32(1): 404-416, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28928244

RESUMEN

Zinc, an essential micronutrient, has a cancer preventive role. Zinc deficiency has been shown to contribute to the progression of esophageal cancer. Orai1, a store-operated Ca2+ entry (SOCE) channel, was previously reported to be highly expressed in tumor tissues removed from patients with esophageal squamous cell carcinoma (ESCC) with poor prognosis, and elevation of its expression contributes to both hyperactive intracellular Ca2+ oscillations and fast cell proliferation in human ESCC cells. However, the molecular basis of cancer preventive functions of zinc and its association with Orai1-mediated cell proliferation remains unknown. The present study shows that zinc supplementation significantly inhibits proliferation of ESCC cell lines and that the effect of zinc is reversible with N,N,N',N'-tetrakis (2-pyridylmethyl) ethylenediamine, a specific Zn2+ chelator, whereas nontumorigenic esophageal epithelial cells are significantly less sensitive to zinc treatment. Fluorescence live cell imaging revealed that extracellular Zn2+ exerted rapid inhibitory effects on Orai1-mediated SOCE and on intracellular Ca2+ oscillations in the ESCC cells. Knockdown of Orai1 or expression of Orai1 mutants with compromised zinc binding significantly diminished sensitivity of the cancer cells to zinc treatment in both SOCE and cell proliferation analyses. These data suggest that zinc may inhibit cell proliferation of esophageal cancer cells through Orai1-mediated intracellular Ca2+ oscillations and reveal a possible molecular basis for zinc-induced cancer prevention and Orai1-SOCE signaling pathway in cancer cells.-Choi, S., Cui, C., Luo, Y., Kim, S.-H., Ko, J.-K., Huo, X., Ma, J., Fu, L.-W., Souza, R. F., Korichneva, I., Pan, Z. Selective inhibitory effects of zinc on cell proliferation in esophageal squamous cell carcinoma through Orai1.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Proteína ORAI1/metabolismo , Zinc/farmacología , Sustitución de Aminoácidos , Señalización del Calcio/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quelantes/farmacología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Etilenodiaminas/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteína ORAI1/antagonistas & inhibidores , Proteína ORAI1/genética
12.
Acta Pharmacol Sin ; 39(7): 1120-1132, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29926844

RESUMEN

Zinc is an essential nutrient for human health and has anti-oxidative stress and anti-inflammatory functions. The association between zinc deficiency and the development of cardiovascular diseases (CVDs) has been supported by numerous studies. Supplementing zinc can reduce the risk of atherosclerosis and protect against myocardial infarction and ischemia/reperfusion injury. In this review we summarize the evidence in the literature, to consolidate the current knowledge on the dysregulation of zinc homeostasis in CVDs, and to explore the significant roles of the zinc homeostasis-regulatory proteins in cardiac physiology and pathophysiology. Moreover, this review also deliberates on the potential diagnostic and prognostic implications of zinc/zinc homeostasis-associated molecules (ZIP, ZnT, and MTs) in CVDs.


Asunto(s)
Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/metabolismo , Estrés Oxidativo , Zinc/deficiencia , Zinc/metabolismo , Animales , Homeostasis , Humanos
13.
Int J Mol Sci ; 19(4)2018 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-29621141

RESUMEN

Chronic heart failure (CHF) is a major contributor to cardiovascular disease and is the leading cause of hospitalization for those over the age of 65, which is estimated to account for close to seventy billion dollars in healthcare costs by 2030 in the US alone. The successful therapies for preventing and reversing CHF progression are urgently required. One strategy under active investigation is to restore dysregulated myocardial calcium (Ca2+), a hallmark of CHF. It is well established that intracellular Ca2+ concentrations are tightly regulated to control efficient myocardial systolic contraction and diastolic relaxation. Among the many cell surface proteins and intracellular organelles that act as the warp and woof of the regulatory network controlling intracellular Ca2+ signals in cardiomyocytes, sarco/endoplasmic reticulum Ca2+ ATPase type 2a (SERCA2a) undoubtedly plays a central role. SERCA2a is responsible for sequestrating cytosolic Ca2+ back into the sarcoplasmic reticulum during diastole, allowing for efficient uncoupling of actin-myosin and subsequent ventricular relaxation. Accumulating evidence has demonstrated that the expression of SERCA2a is downregulated in CHF, which subsequently contributes to severe systolic and diastolic dysfunction. Therefore, restoring SERCA2a expression and improving cardiomyocyte Ca2+ handling provides an excellent alternative to currently used transplantation and mechanical assist devices in the treatment of CHF. Indeed, advancements in safe and effective gene delivery techniques have led to the emergence of SERCA2a gene therapy as a potential therapeutic choice for CHF patients. This mini-review will succinctly detail the progression of SERCA2a gene therapy from its inception in plasmid and animal models, to its clinical trials in CHF patients, highlighting potential avenues for future work along the way.


Asunto(s)
Calcio/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/terapia , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Dependovirus/genética , Terapia Genética/métodos , Humanos
14.
J Biol Chem ; 290(22): 13830-9, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25869134

RESUMEN

Zinc is an essential trace element that participates in a wide range of biological functions, including wound healing. Although Zn(2+) deficiency has been linked to compromised wound healing and tissue repair in human diseases, the molecular mechanisms underlying Zn(2+)-mediated tissue repair remain unknown. Our previous studies established that MG53, a TRIM (tripartite motif) family protein, is an essential component of the cell membrane repair machinery. Domain homology analysis revealed that MG53 contains two Zn(2+)-binding motifs. Here, we show that Zn(2+) binding to MG53 is indispensable to assembly of the cell membrane repair machinery. Live cell imaging illustrated that Zn(2+) entry from extracellular space is essential for translocation of MG53-containing vesicles to the acute membrane injury sites for formation of a repair patch. The effect of Zn(2+) on membrane repair is abolished in mg53(-/-) muscle fibers, suggesting that MG53 functions as a potential target for Zn(2+) during membrane repair. Mutagenesis studies suggested that both RING and B-box motifs of MG53 constitute Zn(2+)-binding domains that contribute to MG53-mediated membrane repair. Overall, this study establishes a base for Zn(2+) interaction with MG53 in protection against injury to the cell membrane.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Zinc/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Reparación del ADN , Electrodos , Escherichia coli/metabolismo , Humanos , Proteínas de la Membrana , Ratones , Ratones Transgénicos , Microscopía Confocal , Músculo Esquelético/metabolismo , Mutación , Oxidación-Reducción , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Medicina Regenerativa , Transducción de Señal , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo , Cicatrización de Heridas
15.
Am J Physiol Renal Physiol ; 311(2): F320-9, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226107

RESUMEN

Human organic anion transporter 1 (hOAT1), expressed at the basolateral membrane of kidney proximal tubule cells, mediates the active renal secretion of a diverse array of clinically important drugs, including anti-human immunodeficiency virus therapeutics, antitumor drugs, antibiotics, antihypertensives, and anti-inflammatories. We have previously demonstrated that posttranslational modification of hOAT1 by ubiquitination is an important mechanism for the regulation of this transporter. The present study aimed at identifying the ubiquitin ligase for hOAT1 and its mechanism of action. We showed that overexpression of neural precursor cell expressed, developmentally downregulated (Nedd)4-1, an E3 ubiquitin ligase, enhanced hOAT1 ubiquitination, decreased hOAT1 expression at the cell surface, and inhibited hOAT1 transport activity. In contrast, overexpression of the ubiquitin ligase-dead mutant Nedd4-1/C867S was without effects on hOAT1. Furthermore, knockdown of endogenously expressed Nedd4-1 by Nedd4-1-specific small interfering RNA reduced hOAT1 ubiquitination. Immunoprecipitation experiments in cultured cells and rat kidney slices and immunofluorescence experiments in rat kidney slices showed that there was a physical interaction between OAT1 and Nedd4-1. Nedd4-1 contains four protein-protein interacting WW domains. When these WW domains were inactivated by mutating two amino acid residues in each of the four WW domains (Mut-WW1: V210W/H212G, Mut-WW2: V367W/H369G, Mut-WW3: I440W/H442G, and Mut-WW4: I492W/H494G, respectively), only Mut-WW2 and Mut-WW3 significantly lost their ability to bind and to ubiquitinate hOAT1. As a result, Mut-WW2 and Mut-WW3 were unable to suppress hOAT1-mediated transport as effectively as wild-type Nedd4-1. In conclusion, this is the first demonstration that Nedd4-1 regulates hOAT1 ubiquitination, expression, and transport activity through its WW2 and WW3 domains.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Biotinilación , Células COS , Chlorocebus aethiops , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Células HEK293 , Humanos , Técnicas In Vitro , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Mutagénesis Sitio-Dirigida , Ubiquitina-Proteína Ligasas Nedd4 , Proteína 1 de Transporte de Anión Orgánico/genética , Unión Proteica , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
16.
Nature ; 459(7246): 596-600, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19387438

RESUMEN

Ca(2+) mobilization from intracellular stores represents an important cell signalling process that is regulated, in mammalian cells, by inositol-1,4,5-trisphosphate (InsP(3)), cyclic ADP ribose and nicotinic acid adenine dinucleotide phosphate (NAADP). InsP(3) and cyclic ADP ribose cause the release of Ca(2+) from sarcoplasmic/endoplasmic reticulum stores by the activation of InsP(3) and ryanodine receptors (InsP(3)Rs and RyRs). In contrast, the nature of the intracellular stores targeted by NAADP and the molecular identity of the NAADP receptors remain controversial, although evidence indicates that NAADP mobilizes Ca(2+) from lysosome-related acidic compartments. Here we show that two-pore channels (TPCs) comprise a family of NAADP receptors, with human TPC1 (also known as TPCN1) and chicken TPC3 (TPCN3) being expressed on endosomal membranes, and human TPC2 (TPCN2) on lysosomal membranes when expressed in HEK293 cells. Membranes enriched with TPC2 show high affinity NAADP binding, and TPC2 underpins NAADP-induced Ca(2+) release from lysosome-related stores that is subsequently amplified by Ca(2+)-induced Ca(2+) release by InsP(3)Rs. Responses to NAADP were abolished by disrupting the lysosomal proton gradient and by ablating TPC2 expression, but were only attenuated by depleting endoplasmic reticulum Ca(2+) stores or by blocking InsP(3)Rs. Thus, TPCs form NAADP receptors that release Ca(2+) from acidic organelles, which can trigger further Ca(2+) signals via sarcoplasmic/endoplasmic reticulum. TPCs therefore provide new insights into the regulation and organization of Ca(2+) signals in animal cells, and will advance our understanding of the physiological role of NAADP.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , NADP/análogos & derivados , Orgánulos/metabolismo , Animales , Canales de Calcio/genética , Señalización del Calcio/efectos de los fármacos , Línea Celular , Pollos , Humanos , Concentración de Iones de Hidrógeno , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , NADP/metabolismo , NADP/farmacología , Orgánulos/efectos de los fármacos , Unión Proteica
17.
J Neurosci ; 33(9): 3865-78, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23447598

RESUMEN

During corticogenesis, pituitary adenylate cyclase-activating polypeptide (PACAP; ADCYAP1) may contribute to proliferation control by activating PAC1 receptors of neural precursors in the embryonic ventricular zone. PAC1 receptors, specifically the hop and short isoforms, couple differentially to and activate distinct pathways that produce pro- or anti-mitogenic actions. Previously, we found that PACAP was an anti-mitogenic signal from embryonic day 13.5 (E13.5) onward both in culture and in vivo and activated cAMP signaling through the short isoform. However, we now find that mice deficient in PACAP exhibited a decrease in the BrdU labeling index (LI) in E9.5 cortex, suggesting that PACAP normally promotes proliferation at this stage. To further define mechanisms, we established a novel culture model in which the viability of very early cortical precursors (E9.5 mouse and E10.5 rat) could be maintained. At this stage, we found that PACAP evoked intracellular calcium fluxes and increased phospho-PKC levels, as well as stimulated G1 cyclin mRNAs and proteins, S-phase entry, and proliferation without affecting cell survival. Significantly, expression of hop receptor isoform was 24-fold greater than the short isoform at E10.5, a ratio that was reversed at E14.5 when short expression was 15-fold greater and PACAP inhibited mitogenesis. Enhanced hop isoform expression, elicited by in vitro treatment of E10.5 precursors with retinoic acid, correlated with sustained pro-mitogenic action of PACAP beyond the developmental switch. Conversely, depletion of hop receptor using short-hairpin RNA abolished PACAP mitogenic stimulation at E10.5. These observations suggest that PACAP elicits temporally specific effects on cortical proliferation via developmentally regulated expression of specific receptor isoforms.


Asunto(s)
Corteza Cerebral/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Mitógenos/farmacología , Neuronas/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Isoformas de ARN/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Factores de Edad , Animales , Bromodesoxiuridina/metabolismo , Calcio/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , División Celular/efectos de los fármacos , División Celular/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Corteza Cerebral/embriología , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/embriología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Femenino , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Etiquetado Corte-Fin in Situ , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitógenos/deficiencia , Mitógenos/genética , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/deficiencia , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Embarazo , Proteína Quinasa C/metabolismo , Piridinas/farmacología , Pirrolidinonas/farmacología , Isoformas de ARN/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Tretinoina/farmacología
18.
J Biol Chem ; 288(4): 2103-9, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23223241

RESUMEN

Functional coupling between inositol (1,4,5)-trisphosphate receptor (IP(3)R) and ryanodine receptor (RyR) represents a critical component of intracellular Ca(2+) signaling in many excitable cells; however, the role of this mechanism in skeletal muscle remains elusive. In skeletal muscle, RyR-mediated Ca(2+) sparks are suppressed in resting conditions, whereas application of transient osmotic stress can trigger activation of Ca(2+) sparks that are restricted to the periphery of the fiber. Here we show that onset of these spatially confined Ca(2+) sparks involves interaction between activation of IP(3)R and RyR near the sarcolemmal membrane. Pharmacological prevention of IP(3) production or inhibition of IP(3)R channel activity abolishes stress-induced Ca(2+) sparks in skeletal muscle. Although genetic ablation of the type 2 IP(3)R does not appear to affect Ca(2+) sparks in skeletal muscle, specific silencing of the type 1 IP(3)R leads to ablation of stress-induced Ca(2+) sparks. Our data indicate that membrane-delimited signaling involving cross-talk between IP(3)R1 and RyR1 contributes to Ca(2+) spark activation in skeletal muscle.


Asunto(s)
Calcio/metabolismo , Regulación de la Expresión Génica , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Calcio/química , Señalización del Calcio , Ratones , Microscopía Confocal/métodos , Modelos Biológicos , Modelos Genéticos , Ósmosis , Técnicas de Placa-Clamp , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
19.
Methods Mol Biol ; 2816: 69-75, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38977589

RESUMEN

Intracellular Ca2+ can be conveniently monitored by sensitive Ca2+ fluorescent dyes in live cells. The Gαq involved lipid signaling pathways and, thus, can be studied by intracellular Ca2+ imaging. Here we describe the protocols to measure intracellular Ca2+ for studying PEG2-EP1 activity in esophageal smooth muscle cells. The ratiometric Fura-2 imaging provides quantitative data, and the Fluo-4 confocal microscopic imaging has high-spatial resolution.


Asunto(s)
Calcio , Receptores Acoplados a Proteínas G , Calcio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Microscopía Confocal/métodos , Transducción de Señal , Miocitos del Músculo Liso/metabolismo , Señalización del Calcio , Humanos , Xantenos/metabolismo , Fura-2/metabolismo , Metabolismo de los Lípidos , Esófago/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Compuestos de Anilina
20.
ACS Biomater Sci Eng ; 10(6): 3759-3774, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38800901

RESUMEN

Conductive biomaterials offer promising solutions to enhance the maturity of cultured cardiomyocytes. While the conventional culture of cardiomyocytes on nonconductive materials leads to more immature characteristics, conductive microenvironments have the potential to support sarcomere development, gap junction formation, and beating of cardiomyocytes in vitro. In this study, we systematically investigated the behaviors of cardiomyocytes on aligned electrospun fibrous membranes composed of elastic and biodegradable polyurethane (PU) doped with varying concentrations of reduced graphene oxide (rGO). Compared to PU and PU-4%rGO membranes, the PU-10%rGO membrane exhibited the highest conductivity, approaching levels close to those of native heart tissue. The PU-rGO membranes retained anisotropic viscoelastic behavior similar to that of the porcine left ventricle and a superior tensile strength. Neonatal rat cardiomyocytes (NRCMs) and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on the PU-rGO membranes displayed enhanced maturation with cell alignment and enhanced sarcomere structure and gap junction formation with PU-10%rGO having the most improved sarcomere structure and CX-43 presence. hiPSC-CMs on the PU-rGO membranes exhibited a uniform and synchronous beating pattern compared with that on PU membranes. Overall, PU-10%rGO exhibited the best performance for cardiomyocyte maturation. The conductive PU-rGO membranes provide a promising matrix for in vitro cardiomyocyte culture with promoted cell maturation/functionality and the potential for cardiac disease treatment.


Asunto(s)
Grafito , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Poliuretanos , Poliuretanos/química , Poliuretanos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/citología , Grafito/química , Grafito/farmacología , Animales , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratas , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Diferenciación Celular/efectos de los fármacos , Andamios del Tejido/química , Células Cultivadas , Elasticidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA