Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
1.
Clin Transl Med ; 14(5): e1694, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38797942

RESUMEN

BACKGROUND: BRAFV600E is the most common genetic mutation in differentiated thyroid cancer (DTC) occurring in 60% of patients and drives malignant tumour cell phenotypes including proliferation, metastasis and immune-escape. BRAFV600E-mutated papillary thyroid cancer (PTC) also displays greatly reduced expression of thyroid differentiation markers, thus tendency to radioactive iodine (RAI) refractory and poor prognosis. Therefore, understanding the molecular mechanisms and main oncogenic events underlying BRAFV600E will guide future therapy development. METHODS: Bioinformatics and clinical specimen analyses, genetic manipulation of BRAFV600E-induced PTC model, functional and mechanism exploration guided with transcriptomic screening, as well as systematic rescue experiments were applied to investigate miR-31 function within BRAFV600E-induced thyroid cancer development. Besides, nanoparticles carrying miR-31 antagomirs were testified to alleviate 131I iodide therapy on PTC models. RESULTS: We identify miR-31 as a significantly increased onco-miR in BRAFV600E-associated PTC that promotes tumour progression, metastasis and RAI refractoriness via sustained Wnt/ß-catenin signalling. Mechanistically, highly activated BRAF/MAPK pathway induces miR-31 expression via c-Jun-mediated transcriptional regulation across in vitro and transgenic mouse models. MiR-31 in turn facilitates ß-catenin stabilisation via directly repressing tumour suppressors CEBPA and DACH1, which direct the expression of multiple essential Wnt/ß-catenin pathway inhibitors. Genetic functional assays showed that thyroid-specific knockout of miR-31 inhibited BRAFV600E-induced PTC progression, and strikingly, enhanced expression of sodium-iodide symporter and other thyroid differentiation markers, thus promoted 131I uptake. Nanoparticle-mediated application of anti-miR-31 antagomirs markedly elevated radio-sensitivity of BRAFV600E-induced PTC tumours to 131I therapy, and efficiently suppressed tumour progression in the pre-clinical mouse model. CONCLUSIONS: Our findings elucidate a novel BRAF/MAPK-miR-31-Wnt/ß-catenin regulatory mechanism underlying clinically BRAFV600E-associated DTC tumourigenesis and dedifferentiation, also highlight a potential adjuvant therapeutic strategy for advanced DTC.


Asunto(s)
MicroARNs , Proteínas Proto-Oncogénicas B-raf , Neoplasias de la Tiroides , Animales , Humanos , Ratones , Carcinogénesis/genética , Desdiferenciación Celular/genética , Desdiferenciación Celular/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/metabolismo
2.
Phytomedicine ; 130: 155704, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38759316

RESUMEN

BACKGROUND: Dysregulation of vascular smooth muscle cell (VSMC) function leads to a variety of diseases such as atherosclerosis and hyperplasia after injury. However, antiproliferative drug targeting VSMC exhibits poor specificity. Therefore, there is an urgent to develop highly specific antiproliferative drugs to prevention and treatment VSMC dedifferentiation associated arteriosclerosis. Kanglexin (KLX), a new anthraquinone compound designed by our team, has potential to regulate VSMC phenotype according to the physicochemical properties. PURPOSE: This project aims to evaluate the therapeutic role of KLX in VSMC dedifferentiation and atherosclerosis, neointimal formation and illustrates the underlying molecular mechanism. METHODS: In vivo, the ApoE-/- mice were fed with high-fat diet (HFD) for a duration of 13 weeks to establish the atherosclerotic model. And rat carotid artery injury model was performed to establish the neointimal formation model. In vitro, PDGF-BB was used to induce VSMC dedifferentiation. RESULTS: We found that KLX ameliorated the atherosclerotic progression including atherosclerotic lesion formation, lipid deposition and collagen deposition in aorta and aortic sinus in atherosclerotic mouse model. In addition, The administration of KLX effectively ameliorated neointimal formation in the carotid artery following balloon injury in SD rats. The findings derived from molecular docking and surface plasmon resonance (SPR) experiments unequivocally demonstrate that KLX had potential to bind PDGFR-ß. Mechanism research work proved that KLX prevented VSMC proliferation, migration and dedifferentiation via activating the PDGFR-ß-MEK -ERK-ELK-1/KLF4 signaling pathway. CONCLUSION: Collectively, we demonstrated that KLX effectively attenuated the progression of atherosclerosis in ApoE-/- mice and carotid arterial neointimal formation in SD rats by inhibiting VSMC phenotypic conversion via PDGFR-ß-MEK-ERK-ELK-1/KLF4 signaling. KLX exhibits promising potential as a viable therapeutic agent for the treatment of VSMC phenotype conversion associated arteriosclerosis.


Asunto(s)
Desdiferenciación Celular , Factor 4 Similar a Kruppel , Músculo Liso Vascular , Neointima , Ratas Sprague-Dawley , Animales , Desdiferenciación Celular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Masculino , Ratones , Neointima/tratamiento farmacológico , Ratas , Antraquinonas/farmacología , Aterosclerosis/tratamiento farmacológico , Miocitos del Músculo Liso/efectos de los fármacos , Arteriosclerosis/tratamiento farmacológico , Arteriosclerosis/prevención & control , Modelos Animales de Enfermedad , Transducción de Señal/efectos de los fármacos , Dieta Alta en Grasa , Ratones Endogámicos C57BL , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Simulación del Acoplamiento Molecular , Proliferación Celular/efectos de los fármacos , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Becaplermina/farmacología
3.
Tissue Eng Part A ; 30(9-10): 415-425, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38323554

RESUMEN

Expansion of chondrocytes presents a major obstacle in the cartilage regeneration procedure, such as matrix-induced autologous chondrocyte implantation. Dedifferentiation of chondrocytes during the expansion process leads to the emergence of a fibrotic (chondrofibrotic) phenotype that decreases the chondrogenic potential of the implanted cells. We aim to (1) determine the extent that chromatin architecture of H3K27me3 and H3K9me3 remodels during dedifferentiation and persists after the transfer to a three-dimensional (3D) culture; and (2) to prevent this persistent remodeling to enhance the chondrogenic potential of expanded bovine chondrocytes, used as a model system. Chromatin architecture remodeling of H3K27me3 and H3K9me3 was observed at 0 population doublings, 8 population doublings, and 16 population doublings (PD16) in a two-dimensional (2D) culture and after encapsulation of the expanded chondrocytes in a 3D hydrogel culture. Chondrocytes were treated with inhibitors of epigenetic modifiers (epigenetic priming) for PD16 and then encapsulated in 3D hydrogels. Chromatin architecture of chondrocytes and gene expression were evaluated before and after encapsulation. We observed a change in chromatin architecture of epigenetic modifications H3K27me3 and H3K9me3 during chondrocyte dedifferentiation. Although inhibiting enzymes that modify H3K27me3 and H3K9me3 did not alter the dedifferentiation process in 2D culture, applying these treatments during the 2D expansion did increase the expression of select chondrogenic genes and protein deposition of type II collagen when transferred to a 3D environment. Overall, we found that epigenetic priming of expanded bovine chondrocytes alters the cell fate when chondrocytes are later encapsulated into a 3D environment, providing a potential method to enhance the success of cartilage regeneration procedures.


Asunto(s)
Condrocitos , Condrogénesis , Epigénesis Genética , Animales , Condrocitos/metabolismo , Condrocitos/citología , Bovinos , Condrogénesis/efectos de los fármacos , Histonas/metabolismo , Células Cultivadas , Desdiferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos
4.
Am J Respir Cell Mol Biol ; 66(1): 23-37, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34236953

RESUMEN

The U.S. Food and Drug Administration-approved proteasomal inhibitor bortezomib (BTZ) has attracted interest for its potential antifibrotic actions. However, neither its in vivo efficacy in lung fibrosis nor its dependence on proteasome inhibition has been conclusively defined. In this study, we assessed the therapeutic efficacy of BTZ in a mouse model of pulmonary fibrosis, developed an in vitro protocol to define its actions on diverse fibroblast activation parameters, determined its reliance on proteasome inhibition for these actions in vivo and in vitro, and explored alternative mechanisms of action. The therapeutic administration of BTZ diminished the severity of pulmonary fibrosis without reducing proteasome activity in the lung. In experiments designed to mimic this lack of proteasome inhibition in vitro, BTZ reduced fibroblast proliferation, differentiation into myofibroblasts, and collagen synthesis. It promoted dedifferentiation of myofibroblasts and overcame their characteristic resistance to apoptosis. Mechanistically, BTZ inhibited kinases important for fibroblast activation while inducing the expression of DUSP1 (dual-specificity protein phosphatase 1), and knockdown of DUSP1 abolished its antifibrotic actions in fibroblasts. Collectively, these findings suggest that BTZ exhibits a multidimensional profile of robust inhibitory actions on lung fibroblasts as well as antifibrotic actions in vivo. Unexpectedly, these actions appear to be independent of proteasome inhibition, instead attributable to the induction of DUSP1.


Asunto(s)
Bortezomib/uso terapéutico , Fibroblastos/patología , Inhibidores de Proteasoma/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Adulto , Apoptosis/efectos de los fármacos , Bleomicina , Bortezomib/farmacología , Desdiferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Fosfatasa 1 de Especificidad Dual/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/efectos de los fármacos , Humanos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , FN-kappa B/metabolismo , Prostaglandinas/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Receptor fas/metabolismo
5.
Mol Neurobiol ; 59(1): 429-444, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34708329

RESUMEN

Silent information regulator 6 (SIRT6) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Previous studies have been reported a pro-regenerative role of SIRT6 in central nervous system injury. However, the role of SIRT6 in peripheral nerve injury is still unknown. Given the importance and necessity of Schwann cell dedifferentiation response to peripheral nerve injury, we aim to investigate the molecular mechanism of SIRT6 steering Schwann cell dedifferentiation during Wallerian degeneration in injured peripheral nerve. Herein, we first examined the expression pattern of SIRT6 after peripheral nerve injury. Using the explants of sciatic nerve, an ex vivo model of nerve degeneration, we provided evidences indicating that SIRT6 inhibitor accelerates Schwann cell dedifferentiation as well as axonal and myelin degeneration, while SIRT6 activator attenuates this process. Moreover, in an in vitro Schwann cell dedifferentiation model, we found SIRT6 inhibitor promotes Schwann cell dedifferentiation through upregulating the expression of c-Jun. In addition, downregulation of c-Jun reverse the effects of SIRT6 inhibition on the Schwann cells dedifferentiation and axonal and myelin degeneration. In summary, we first described SIRT6 acts as a negative regulator for Schwann cells dedifferentiation during Wallerian degeneration and c-Jun worked as a direct downstream partner of SIRT6 in injured peripheral nerve.


Asunto(s)
Desdiferenciación Celular/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Células de Schwann/metabolismo , Sirtuinas/metabolismo , Degeneración Walleriana/metabolismo , Animales , Desdiferenciación Celular/efectos de los fármacos , Traumatismos de los Nervios Periféricos/patología , Ratas , Células de Schwann/efectos de los fármacos , Sirtuinas/antagonistas & inhibidores , Degeneración Walleriana/patología
6.
J Neuroinflammation ; 18(1): 293, 2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34920725

RESUMEN

BACKGROUND: Lysophosphatidic acid (LPA) is a pleiotropic lipid messenger that addresses at least six specific G-protein coupled receptors. Accumulating evidence indicates a significant involvement of LPA in immune cell regulation as well as Schwann cell physiology, with potential relevance for the pathophysiology of peripheral neuroinflammation. However, the role of LPA signaling in inflammatory neuropathies has remained completely undefined. Given the broad expression of LPA receptors on both Schwann cells and cells of the innate and adaptive immune system, we hypothesized that inhibition of LPA signaling may ameliorate the course of disease in experimental autoimmune neuritis (EAN). METHODS: We induced active EAN by inoculation of myelin protein 2 peptide (P255-78) in female Lewis rats. Animals received the orally available LPA receptor antagonist AM095, specifically targeting the LPA1 receptor subtype. AM095 was administered daily via oral gavage in a therapeutic regimen from 10 until 28 days post-immunization (dpi). Analyses were based on clinical testing, hemogram profiles, immunohistochemistry and morphometric assessment of myelination. RESULTS: Lewis rats treated with AM095 displayed a significant improvement in clinical scores, most notably during the remission phase. Cellular infiltration of sciatic nerve was only discretely affected by AM095. Hemogram profiles indicated no impact on circulating leukocytes. However, sciatic nerve immunohistochemistry revealed a reduction in the number of Schwann cells expressing the dedifferentiation marker Sox2 paralleled by a corresponding increase in differentiating Sox10-positive Schwann cells. In line with this, morphometric analysis of sciatic nerve semi-thin sections identified a significant increase in large-caliber myelinated axons at 28 dpi. Myelin thickness was unaffected by AM095. CONCLUSION: Thus, LPA1 signaling may present a novel therapeutic target for the treatment of inflammatory neuropathies, potentially affecting regenerative responses in the peripheral nerve by modulating Schwann cell differentiation.


Asunto(s)
Desdiferenciación Celular/fisiología , Neuritis Autoinmune Experimental/inmunología , Receptores del Ácido Lisofosfatídico/inmunología , Células de Schwann/inmunología , Transducción de Señal/fisiología , Animales , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/uso terapéutico , Desdiferenciación Celular/efectos de los fármacos , Femenino , Isoxazoles/farmacología , Isoxazoles/uso terapéutico , Neuritis Autoinmune Experimental/tratamiento farmacológico , Neuritis Autoinmune Experimental/metabolismo , Ratas , Ratas Endogámicas Lew , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Oxid Med Cell Longev ; 2021: 3119953, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34900084

RESUMEN

Mangiferin is a naturally occurring xanthone C-glycoside that is widely found in various plants. Previous studies have reported that mangiferin inhibits tumor cell proliferation and migration. Excessive proliferation and migration of vascular smooth muscle cells (SMCs) is associated with neointimal hyperplasia in coronary arteries. However, the role and mechanism of mangiferin action in neointimal hyperplasia is still unknown. In this study, a mouse carotid artery ligation model was established, and primary rat smooth muscle cells were isolated and used for mechanistic assays. We found that mangiferin alleviated neointimal hyperplasia, inhibited proliferation and migration of SMCs, and promoted platelets derive growth factors-BB- (PDGF-BB-) induced contractile phenotype in SMCs. Moreover, mangiferin attenuated neointimal formation by inhibiting mitochondrial fission through the AMPK/Drp1 signaling pathway. These findings suggest that mangiferin has the potential to maintain vascular homeostasis and inhibit neointimal hyperplasia.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Becaplermina/farmacología , Proliferación Celular/efectos de los fármacos , Dinaminas/metabolismo , Xantonas/farmacología , Animales , Arterias Carótidas/patología , Desdiferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Hiperplasia/metabolismo , Hiperplasia/patología , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Dinámicas Mitocondriales/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo
8.
Life Sci ; 287: 120126, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34758295

RESUMEN

AIMS: Astrocytes re-acquire stem cell potential upon inflammation, thereby becoming a promising source of cells for regenerative medicine. Nanog is an essential transcription factor to maintain the characteristics of stem cells. We aimed to investigate the role of Nanog in astrocyte dedifferentiation. MAIN METHODS: TNF-α was used to induce the dedifferentiation of primary rat spinal cord astrocytes. The expression of immature markers CD44 and Musashi-1 was detected by qRT-PCR and immunofluorescence. The Nanog gene is knocked down by small interference RNA. Nanog expression was measured by qRT-PCR and western blotting. BAY 11-7082 was used to suppress NF-κB signals in astrocytes. NF-κB signaling was evaluated by Western blotting. KEY FINDINGS: Our results showed that TNF-α promoted the re-expression of CD44 and Musashi-1 in astrocytes. Dedifferentiated astrocytes could be induced to differentiate into oligodendrocyte lineage cells indicating that the astrocytes had pluripotency. In addition, TNF-α treatment activated NF-κB signaling pathway and up-regulated Nanog. Knockdown of Nanog reversed the increase of CD44 and Musashi-1 induced by TNF-α without affecting the activation of NF-κB signaling. Importantly, blocking NF-κB signaling by BAY 11-7082 inhibited the expression of immature markers suggesting that TNF-α induces dedifferentiation of astrocytes through the NF-κB signaling pathway. BAY 11-7082 could also inhibit the expression of Nanog, which indicated that Nanog was regulated by NF-κB signaling pathway. SIGNIFICANCE: These findings indicate that activation of the NF-κB signaling pathway through TNF-α leads to astrocytes dedifferentiation via Nanog. These results expand our understanding of the mechanism of astrocytes dedifferentiation.


Asunto(s)
Astrocitos/metabolismo , Desdiferenciación Celular/fisiología , FN-kappa B/metabolismo , Proteína Homeótica Nanog/biosíntesis , Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Astrocitos/efectos de los fármacos , Desdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
9.
J Mater Sci Mater Med ; 32(9): 121, 2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-34499229

RESUMEN

Cardiovascular diseases are the leading cause of death in the world, cell therapies have been shown to recover cardiac function in animal models. Biomaterials used as scaffolds can solve some of the problems that cell therapies currently have, plasma polymerized pyrrole (PPPy) is a biomaterial that has been shown to promote cell adhesion and survival. The present research aimed to study PPPy nanoparticles (PPPyN) interaction with adult rat ventricular cardiomyocytes (ARVC), to explore whether PPPyN could be employed as a nanoscaffold and develop cardiac microtissues. PPPyN with a mean diameter of 330 nm were obtained, the infrared spectrum showed that some pyrrole rings are fragmented and that some fragments of the ring can be dehydrogenated during plasma synthesis, it also showed the presence of amino groups in the structure of PPPyN. PPPyN had a significant impact on the ARVC´s shape, delaying dedifferentiation, necrosis, and apoptosis processes, moreover, the cardiomyocytes formed cell aggregates up to 1.12 mm2 with some aligned cardiomyocytes and generated fibers on its surface similar to cardiac extracellular matrix. PPPyN served as a scaffold for adult ARVC. Our results indicate that PPPyN-scaffold is a biomaterial that could have potential application in cardiac cell therapy (CCT).


Asunto(s)
Miocitos Cardíacos/efectos de los fármacos , Nanopartículas/química , Pirroles/farmacología , Animales , Desdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Matriz Extracelular/química , Matriz Extracelular/efectos de los fármacos , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Masculino , Ensayo de Materiales , Miocitos Cardíacos/fisiología , Gases em Plasma/farmacología , Polimerizacion/efectos de los fármacos , Pirroles/química , Ratas , Ratas Wistar
10.
Int J Cancer ; 149(11): 1961-1972, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34469585

RESUMEN

Adipocyte-rich omentum offers "good soil" for disseminating ovarian cancer (OvCa), contributing to therapeutic difficulty. However, little is understood about the association between adipocytes and tumor growth at peritoneal dissemination site. Herein, we report the induction of adipocyte dedifferentiation by OvCa cells and pro-tumorigenic effects of resulted adipocyte-derived fibroblasts. We confirmed that malignant ascites promoted the dedifferentiation of the primary human adipocytes obtained from surgical omental specimen into omental adipocyte-derived fibroblast (O-ADF) that possess both mesenchymal stem cell and myofibroblast-like features. This promotion of dedifferentiation by malignant ascites was blocked by addition of Wnt signaling inhibitor. The effects of dedifferentiated adipocytes in proliferation and migration of OvCa cells were analyzed with in vitro coculturing experimental models and in vivo mice model, and we demonstrated that OvCa cell lines showed enhanced proliferative characteristics, as well as increased migratory abilities upon coculturing with O-ADF. Additionally, exogenous transforming growth factor-ß1 augmented desmoplastic morphological change of O-ADF, leading to higher proliferative ability. Our results suggest that OvCa cells promote dedifferentiation of peritoneal adipocytes by activating Wnt/ß-catenin signaling, and generated O-ADFs exhibit pro-tumoral hallmarks.


Asunto(s)
Adipocitos/patología , Fibroblastos Asociados al Cáncer/patología , Epiplón/patología , Neoplasias Ováricas/patología , Microambiente Tumoral , Células 3T3-L1 , Actinas/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Ascitis/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Desdiferenciación Celular/efectos de los fármacos , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Imidas/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Epiplón/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/secundario , Quinolinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt3A/metabolismo
11.
Signal Transduct Target Ther ; 6(1): 333, 2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-34482361

RESUMEN

Application of differentiation therapy targeting cellular plasticity for the treatment of solid malignancies has been lagging. Nasopharyngeal carcinoma (NPC) is a distinctive cancer with poor differentiation and high prevalence of Epstein-Barr virus (EBV) infection. Here, we show that the expression of EBV latent protein LMP1 induces dedifferentiated and stem-like status with high plasticity through the transcriptional inhibition of CEBPA. Mechanistically, LMP1 upregulates STAT5A and recruits HDAC1/2 to the CEBPA locus to reduce its histone acetylation. HDAC inhibition restored CEBPA expression, reversing cellular dedifferentiation and stem-like status in mouse xenograft models. These findings provide a novel mechanistic epigenetic-based insight into virus-induced cellular plasticity and propose a promising concept of differentiation therapy in solid tumor by using HDAC inhibitors to target cellular plasticity.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Inhibidores de Histona Desacetilasas/farmacología , Carcinoma Nasofaríngeo/tratamiento farmacológico , Factor de Transcripción STAT5/genética , Proteínas de la Matriz Viral/genética , Animales , Desdiferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Plasticidad de la Célula/efectos de los fármacos , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/patogenicidad , Xenoinjertos , Humanos , Ratones , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patología , Carcinoma Nasofaríngeo/virología
12.
J Dermatol Sci ; 103(2): 101-108, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34315630

RESUMEN

BACKGROUND: Systemic sclerosis (SSc) causes progressive fibrosis of multiple organs with the low efficacy of immunosuppressive therapies. Our previous study indicated the SSc pathological pathways are closely correlated with Ca2+ signals, and blockage of the intracellular Ca2+ elevation facilitates inhibition of SSc pathogenesis. OBJECTIVE: Transforming growth factor ß (TGF-ß)-modulated SMAD signaling is crucial in regulating SSc pathogenesis. Whether Ca2+ signals are involved in TGF-ß1/SMAD signaling-induced fibrotic process has been further investigated. METHODS: We utilized TGF-ß1-induced myofibroblasts as a model to detect how Ca2+ signals affected SSc pathogenesis, and investigated the combination of treatment with store-operated Ca2+ entry (SOCE) associated inhibitors, 2-aminoethyl diphenylborinate (2-APB) and SKF96365 to restrain the increased Ca2+ signaling in myofibroblasts. In addition, the SSc bleomycin mouse model was used to detect the effect of 2-APB on SSc pathogenesis in vivo. RESULTS: Our findings revealed increased levels of TGF-ß1 production in SSc was associated with intracellular Ca2+ activity, and inhibition of intracellular Ca2+ regulation by 2-APB resulted in the dedifferentiation of TGF-ß1-induced myofibroblasts. This was due to the fact that 2-APB restrained the expression fibrotic markers, α-SMA, fibronectin and vimentin through inhibiting TGF-ß1/SMAD3 signaling. Thus, subcutaneous injection of 2-APB improved bleomycin-induced skin and pulmonary fibrosis. CONCLUSION: 2-APB is a potential candidate for treating fibrosis, by disrupting intracellular Ca2+ regulation in SSc to induce the dedifferentiation of myofibroblasts and meliorates fibrosis pathogenesis via inhibiting TGF-ß1/SMAD3 signaling.


Asunto(s)
Compuestos de Boro/uso terapéutico , Señalización del Calcio/efectos de los fármacos , Desdiferenciación Celular/efectos de los fármacos , Fibrosis Pulmonar/prevención & control , Esclerodermia Sistémica/prevención & control , Adulto , Anciano , Animales , Bleomicina , Compuestos de Boro/farmacología , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fibrosis Pulmonar/metabolismo , Esclerodermia Sistémica/metabolismo , Adulto Joven
13.
J Clin Invest ; 131(12)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33914706

RESUMEN

Melanoma dedifferentiation has been reported to be a state of cellular resistance to targeted therapies and immunotherapies as cancer cells revert to a more primitive cellular phenotype. Here, we show that, counterintuitively, the biopsies of patient tumors that responded to anti-programmed cell death 1 (anti-PD-1) therapy had decreased expression of melanocytic markers and increased neural crest markers, suggesting treatment-induced dedifferentiation. When modeling the effects in vitro, we documented that melanoma cell lines that were originally differentiated underwent a process of neural crest dedifferentiation when continuously exposed to IFN-γ, through global chromatin landscape changes that led to enrichment in specific hyperaccessible chromatin regions. The IFN-γ-induced dedifferentiation signature corresponded with improved outcomes in patients with melanoma, challenging the notion that neural crest dedifferentiation is entirely an adverse phenotype.


Asunto(s)
Biomarcadores de Tumor , Desdiferenciación Celular/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Interferón gamma/metabolismo , Melanoma , Proteínas de Neoplasias , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo
14.
Mol Med Rep ; 23(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33760132

RESUMEN

Hair loss, including alopecia, is a common and distressing problem for men and women, and as a result, there is considerable interest in developing treatments that can prevent or reverse hair loss. Dermal papillae closely interact with epidermal cells and play a key role during hair follicle induction and hair morphogenesis. As dermal papilla cells (DPCs) lose their hair­inducing ability in monolayer cultures in vitro, it is difficult to obtain de novo hair follicle structures following DPC transplantation in vivo. The present study aimed to explore culture conditions to maintain DPC characteristics using conditioned media (CM) from the supernatant of cultured HaCaT keratinocyte cells supplemented with other components. Initially, it was observed that during passaging of in vitro monolayer DPC cultures, the Wnt/ß­catenin pathway was repressed, while the TGF­ß/Smad pathway was activated, and that HaCaT cells cultivated in 1% fetal bovine serum had higher levels of expression of Wnt3a and Wnt10b compared with normal keratinocytes. Culturing of high­passage (P7) DPCs in CM from HaCaT cells (HaCaT­CM) actively stimulated cell proliferation and maintained Sox2 and Versican expression levels. Supplementation of HaCaT­CM with SB431542 (SB, a TGF­ß receptor inhibitor), CHIR99021, (CHIR, a GSK3α/ß inhibitor and activator of Wnt signaling) and platelet­derived growth factor (PDGF)­AA further increased the expression levels of Sox2, Versican and alkaline phosphatase (ALP) in P7 DPCs. Three­dimensional culture of P7 DPCs using hanging drop cultures in HaCaT­CM supplemented with SB, CHIR and PDGF­AA resulted in larger cell aggregates and a further significant upregulation of Sox2, ALP and Versican expression levels. Taken together, these findings demonstrated that HaCaT­CM supplemented with SB, CHIR and PDGF­AA may preserve the hair­inducing ability of high­passage DPCs and may therefore be useful in reconstructing new hair follicles in vivo.


Asunto(s)
Alopecia/genética , Desdiferenciación Celular/efectos de los fármacos , Dermis/crecimiento & desarrollo , Factor de Crecimiento Derivado de Plaquetas/genética , Alopecia/tratamiento farmacológico , Alopecia/patología , Benzamidas/farmacología , Técnicas de Cultivo de Célula , Medios de Cultivo Condicionados , Dermis/citología , Dioxoles/farmacología , Cabello/crecimiento & desarrollo , Cabello/metabolismo , Folículo Piloso/citología , Folículo Piloso/crecimiento & desarrollo , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Vía de Señalización Wnt/efectos de los fármacos
16.
Neuropharmacology ; 188: 108514, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33684416

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) can lead to discontinuation of chemotherapy and is consequently a serious impediment to effective cancer treatment. Due to our limited understanding of mechanisms underlying the pathogenesis of CIPN, no causal therapy has been approved for relief of this condition. We previously demonstrated that taxanes (paclitaxel and docetaxel) induce Schwann cell dedifferentiation, characterized by increased expression of p75 and galectin-3, ultimately leading to demyelination. These changes appear to be responsible for CIPN pathogenesis. This study was designed to identify a novel candidate therapeutic for CIPN with the ability to suppress paclitaxel-induced Schwann cell dedifferentiation. Given that elevation of cyclic adenosine monophosphate (cAMP) signaling participates in Schwann cell differentiation, we performed immunocytochemical screening of phosphodiesterase (PDE) inhibitors. We found that the PDE3 inhibitor cilostazol strongly promoted differentiation of primary cultures of rat Schwann cells via a mechanism involving cAMP/exchange protein directly activated by cAMP (Epac) signaling. Co-treatment with cilostazol prevented paclitaxel-induced dedifferentiation of Schwann cell cultures and demyelination in a mixed culture of Schwann cells and dorsal root ganglia neurons. Notably, continuous oral administration of cilostazol suppressed Schwann cell dedifferentiation within the sciatic nerve and the development of mechanical hypersensitivity in a mouse model of paclitaxel-related CIPN. Importantly, cilostazol potentiated, rather than inhibited, the anti-cancer effect of paclitaxel on the human breast cancer cell line MDA-MB-231. These findings highlight the potential utility of cilostazol as a causal therapeutic that avoids the development of paclitaxel-related CIPN without compromising anti-cancer properties.


Asunto(s)
Desdiferenciación Celular/efectos de los fármacos , Cilostazol/farmacología , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Células de Schwann/efectos de los fármacos , Animales , Proteínas Sanguíneas , Neoplasias de la Mama , Línea Celular Tumoral , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Femenino , Galectinas , Ganglios Espinales/metabolismo , Humanos , Hiperalgesia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Células de Schwann/metabolismo , Nervio Ciático/metabolismo
17.
Int J Mol Sci ; 22(3)2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525349

RESUMEN

Osteoarthritis (OA) is a degenerative joint disease that affects a lot of people worldwide. Current treatment for OA mainly focuses on halting or slowing down the disease progress and to improve the patient's quality of life and functionality. Autologous chondrocyte implantation (ACI) is a new treatment modality with the potential to promote regeneration of worn cartilage. Traditionally, foetal bovine serum (FBS) is used to expand the chondrocytes. However, the use of FBS is not ideal for the expansion of cells mean for clinical applications as it possesses the risk of animal pathogen transmission and animal protein transfer to host. Human platelet lysate (HPL) appears to be a suitable alternative to FBS as it is rich in biological factors that enhance cell proliferation. Thus far, HPL has been found to be superior in promoting chondrocyte proliferation compared to FBS. However, both HPL and FBS cannot prevent chondrocyte dedifferentiation. Discrepant results have been reported for the maintenance of chondrocyte redifferentiation potential by HPL. These differences are likely due to the diversity in the HPL preparation methods. In the future, more studies on HPL need to be performed to develop a standardized technique which is capable of producing HPL that can maintain the chondrocyte redifferentiation potential reproducibly. This review discusses the in vitro expansion of chondrocytes with FBS and HPL, focusing on its capability to promote the proliferation and maintain the chondrogenic characteristics of chondrocytes.


Asunto(s)
Plaquetas/química , Extractos Celulares/farmacología , Condrocitos/efectos de los fármacos , Medios de Cultivo/farmacología , Osteoartritis/terapia , Albúmina Sérica Bovina/farmacología , Animales , Cartílago/metabolismo , Cartílago/patología , Bovinos , Técnicas de Cultivo de Célula , Desdiferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Extractos Celulares/química , Proliferación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/metabolismo , Condrocitos/trasplante , Condrogénesis/fisiología , Medios de Cultivo/química , Progresión de la Enfermedad , Humanos , Osteoartritis/metabolismo , Osteoartritis/patología , Albúmina Sérica Bovina/aislamiento & purificación , Trasplante Autólogo/métodos
18.
Sci Rep ; 11(1): 1852, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33473142

RESUMEN

The DNA demethylating agent, 5-Azacytidine (5Aza), and histone deacetylase inhibitor, valproic acid (Vpa), can improve the reprogramming efficiencies of pluripotent cells. This study aimed to examine the roles of 5Aza and Vpa in the dedifferentiation of epithelial cell rests of Malassez (ERM) into stem-like cells. Additionally, the ability of stem-like cells to differentiate into mesenchymal cells was evaluated. ERM was cultured in embryonic stem cell medium (ESCM) with 1 µM of 5Aza, or 2 mM of Vpa, or a combination of 5Aza and Vpa. The cells stimulated with both 5Aza and Vpa were named as progenitor-dedifferentiated into stem-like cells (Pro-DSLCs). The Pro-DSLCs cultured in ESCM alone for another week were named as DSLCs. The stem cell markers were significantly higher in the DSLCs than the controls (no additions). The mRNA and protein levels of the endothelial, mesenchymal stem, and osteogenic cell markers were significantly higher in the Pro-DSLCs and DSLCs than the controls. The combination of a demethylating agent and a deacetylated inhibitor induced the dedifferentiation of ERM into DSLCs. The Pro-DSLCs derived from ERM can be directly reprogrammed into mesenchymal-like cells without dedifferentiation into stem-like cells. Isolated ERM treated with epigenetic agents may be used for periodontal regeneration.


Asunto(s)
Azacitidina/farmacología , Reprogramación Celular/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Desdiferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo/química , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Porcinos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Cell Signal ; 80: 109911, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33422645

RESUMEN

Our previous data indicate that both insulin and IGF-1 signallings dysfunction promotes the dedifferentiation of primary human and mouse white adipocytes. Based on the fact that insulin activates mTOR and inhibits autophagy, and autophagy deficiency can inhibit the differentiation of white adipocytes, we speculate that autophagy may be related to the dedifferentiation of white adipocytes. We investigated the underlying mechanism of autophagy during dedifferentiation of mouse 3T3-L1 adipocytes. After incomplete inhibition of insulin and IGF-1 signallings, 3T3-L1 adipocytes manifest dedifferentiation accompanied with an increase of autophagy level. If induction only of autophagy in the adipocytes, then the cells also occur somewhat dedifferentiation, and with a slight decrease of insulin signal, while its degree was weaker than insulin signal inhibited cells. Notably, after inhibition of the insulin and IGF-1 signallings and simultaneously inducing autophagy, the dedifferentiation of 3T3-L1 adipocytes was the most obvious compared with other groups, and the insulin and IGF-1 signallings decreases was greater than the cells with inhibition only of insulin signalling. If inhibition of both insulin signal and autophagy simultaneously, the dedifferentiation of the adipocytes reveals similar tendencies to the cells that insulin signal was inhibited. No significant dedifferentiation occurs of 3T3-L1 cells if only inhibition of autophagy. Taken all together, in this study, we proved that autophagy is positively related to the dedifferentiation of 3T3-L1 adipocytes and is regulated through the insulin-PI3K-AKT-mTOCR1-autophagy pathway. Autophagy may also has a certain degree of negative feedback affect on the insulin signalling of 3T3-L1 cells. Our work may help to better understand the biological properties of mature adipocytes and may help formulate anti-obesity strategies by regulating insulin and insulin signaling level.


Asunto(s)
Autofagia , Desdiferenciación Celular , Insulina/metabolismo , Transducción de Señal , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Autofagia/efectos de los fármacos , Desdiferenciación Celular/efectos de los fármacos , Imidazoles/farmacología , Insulina/química , Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Macrólidos/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología
20.
Curr Opin Immunol ; 70: 15-26, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33429228

RESUMEN

Sustained transcriptional activation of the aryl hydrocarbon receptor (AhR) promotes tumour growth and impairs the immune defence, at least for cutaneous melanoma and glioma. AhR ligands are produced by the tumour microenvironment (TME) and by the tumour itself (intracrine). The recent identification of interleukin-4-induced-1 (IL4I1), a parallel pathway to indoleamine 2 3-dioxygenase 1 (IDO1)/ tryptophan 2,3-dioxygenase (TDO), and its ability to generate AhR ligands, confirms that a complete inhibition of AhR ligand production might be difficult to reach. Here, we have focused on recent discoveries explaining the large varieties of AhR ligands and the functional consequences in terms of cancer cell plasticity and consecutive therapy resistance. We also examined therapeutic strategies targeting the AhR signalling pathway and their possible adverse effects. Since the end of 2019, two phase I clinical trials have investigated the ability of the AhR antagonist to 'reset' the immune system and re-sensitize the cancer cells to therapies by preventing their dedifferentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Neoplasias/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Desdiferenciación Celular/efectos de los fármacos , Desdiferenciación Celular/inmunología , Humanos , Ligandos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA