Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Carcinog ; 61(11): 1043-1055, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36102200

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a common human malignancy characterized by late-stage diagnosis, metastasis, and poor prognosis. Cisplatin (DDP)-based chemotherapy has been the most predominant treatment for patients with ESCC. However, the high rate of DDP resistance and toxicity seriously hinder its clinical application. Then, the optimized strategy and mechanisms for ESCC to enhance DDP sensitivity are in great demand. Accumulating evidence have shown that chaperone proteins are closely related to the tumorigenesis and drug resistance of cancers. Chaperonin containing TCP1 complex 4 (CCT4) is a recent identified member of the family. However, its expression and function in ESCC have not been well illustrated. In this study, we found that CCT4 was highly expressed in human ESCC tissues and cell lines, and closely related to the poor prognosis. Moreover, CCT4 silence raised oxidative stress and inhibited glycolysis of ESCC cells, which significantly inhibited cell proliferation and migration, promoted apoptosis and caused cell cycle arrest in ESCC cells. Interestingly, CCT4 knockdown enhanced the sensitivity of KYSE150 cells to DDP by regulating AMPK/AKT/Nrf2 signaling pathway and inhibiting glycolysis ability. Taken together, our results indicate that targeting CCT4 may be a therapeutic target in ESCC patients, which provides a theoretical basis to enhance the sensitivity of DDP in ESCC.


Asunto(s)
Carcinoma de Células Escamosas , Chaperonina con TCP-1 , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Proteínas Quinasas Activadas por AMP/metabolismo , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Chaperonina con TCP-1/genética , Chaperoninas/metabolismo , Chaperoninas/uso terapéutico , Cisplatino/farmacología , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Mol Carcinog ; 60(7): 440-454, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34003522

RESUMEN

Aberrant expression of kinesin family member 4A (KIF4A), which is associated with tumor progression, has been reported in several types of cancer. However, its expression and the underlying molecular mechanisms regulating the transcription of KIF4A in esophageal squamous cell carcinoma (ESCC) remain largely unclear. Here, we found that high KIF4A expression was positively correlated with tumor stage and poor prognosis in ESCC patients. KIF4A silencing significantly inhibited the growth and migration of ESCC cells, arrested cell cycle, and induced apoptosis. Interestingly, KIF4A expression was positively related to the expression of YAP in human ESCC tissues. YAP knockdown or disrupting YAP/TEAD4 interaction by verteporfin repressed KIF4A expression. Also, KIF4A knockdown significantly inhibited the cell growth induced by YAP overexpression. Mechanistically, YAP activated KIF4A transcriptional expression by TEAD4-mediated direct binding to KIF4A promoter. Finally, KIF4A knockdown and verteporfin treatment synergistically inhibited tumor growth in xenograft models. Together, these results indicated that KIF4A, a novel target gene of YAP/TEAD4, may be a progression and prognostic biomarker of ESCC. Targeting drugs for KIF4A combined with YAP inhibitor may be a novel therapeutic strategy for ESCC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Unión al ADN/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Cinesinas/genética , Proteínas Musculares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anciano , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/genética , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/mortalidad , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/mortalidad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Cinesinas/metabolismo , Masculino , Ratones Desnudos , Persona de Mediana Edad , Proteínas Musculares/genética , Pronóstico , Factores de Transcripción de Dominio TEA , Verteporfina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
3.
Biogerontology ; 22(2): 165-187, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33502634

RESUMEN

Aging is a physiological process mediated by numerous biological and genetic pathways, which are directly linked to lifespan and are a driving force for all age-related diseases. Human life expectancy has greatly increased in the past few decades, but this has not been accompanied by a similar increase in their healthspan. At present, research on aging biology has focused on elucidating the biochemical and genetic pathways that contribute to aging over time. Several aging mechanisms have been identified, primarily including genomic instability, telomere shortening, and cellular senescence. Aging is a driving factor of various age-related diseases, including neurodegenerative diseases, cardiovascular diseases, cancer, immune system disorders, and musculoskeletal disorders. Efforts to find drugs that improve the healthspan by targeting the pathogenesis of aging have now become a hot topic in this field. In the present review, the status of aging research and the development of potential drugs for aging-related diseases, such as metformin, rapamycin, resveratrol, senolytics, as well as caloric restriction, are summarized. The feasibility, side effects, and future potential of these treatments are also discussed, which will provide a basis to develop novel anti-aging therapeutics for improving the healthspan and preventing aging-related diseases.


Asunto(s)
Envejecimiento , Longevidad , Restricción Calórica , Senescencia Celular , Humanos , Esperanza de Vida
4.
Artículo en Inglés | MEDLINE | ID: mdl-38935462

RESUMEN

Osteoarthritis (OA) is a progressive joint disorder characterized by sustained oxidative stress, chronic inflammation, and the degradation of cartilage. Despite extensive research on nanocarrier treatment strategies, the therapeutic efficacy remains limited due to the lack of satisfactory vehicles that can simultaneously exhibit excellent ROS scavenging capabilities and high drug loading capacity for effective nonsurgical management of OA. In this work, we propose an innovative strategy utilizing hollow mesoporous cerium oxide nanospheres coated with membranes derived from apoptotic chondrocytes as a reactive oxygen species "sweeper" for targeted and anti-inflammatory therapy of OA. The developed DEX@HMCeNs@M demonstrates superior drug loading capacity, notable antioxidant properties, favorable biocompatibility, and controlled drug release. By leveraging the camouflage provided by apoptotic chondrocyte membranes, the engineered DEX@HMCeNs@M, which bear natural "eat me" signals, can effectively mimic chondrocyte apoptotic bodies within the joints, thereby enabling targeted delivery of the anti-inflammatory drug DEX and subsequent controlled release triggered by the acidic environment of OA. Both in vitro and in vivo experiments validate the enhanced therapeutic efficacy of our DEX@HMCeNs@M sweeper, which operates through a synergistic mechanism involving scavenging of ROS overproduction, inhibition of inflammation, restoration of mitochondrial damage, and reduction of chondrocyte apoptosis. These findings underscore the potential and efficiency of our developed DEX@HMCeNs@M strategy as an encouraging interventional approach for the progressive treatment of OA.

5.
Bioeng Transl Med ; 8(2): e10414, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36925715

RESUMEN

Bone injury caused changes to surrounding tissues, leading to a large number of osteoclasts appeared to clear the damaged bone tissue before bone regeneration. However, overactive osteoclasts will inhibit bone formation. In this study, we prepared methacrylylated gelatin (GelMA)-based hydrogel to co-crosslink with OP3-4 peptide, a receptor activator of NF-κB ligand (RANKL) binding agent, to achieve the slow release of OP3-4 peptide to inhibit the activation of osteoclasts, thus preventing the long-term existence of osteoclasts from affecting bone regeneration, and promoting osteogenic differentiation. Moreover, CXCL9 secreted by osteoblasts will bind to endogenous VEGF and inhibit vascularization, finally hinder bone formation. Thus, anti-CXCL9 antibodies (A-CXCL9) were also loaded in the hydrogel to neutralize excess CXCL9. The hydrogel slow released of OP3-4 cyclic peptide and A-CXCL9 to simultaneously inhibiting osteoclast activation and promoting vascularization, thereby accelerating the healing of femur defect. Further analysis of osteogenic protein expression and signal pathways showed that the hydrogel may be through activating the AKT-RUNX2-ALP pathway and ultimately promote osteogenic differentiation. This dual-acting hydrogel can effectively prevent nonunion caused by low vascularization and provide long-term support for the treatment of bone injury.

6.
Sci Rep ; 12(1): 6692, 2022 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-35461324

RESUMEN

The crosstalk between osteosarcoma (OS) development and abnormally expressed microRNA (miR)-601 is not explored explicitly. Here, we identified the downregulated miR-601 in osteosarcoma (OS) through a comprehensive bioinformatics analysis of GEO Datasets. The results indicated that miR-601 was downregulated in both OS cells and tissues. The OS patients with reduced expression of miR-601 displayed worse prognosis. The results of in vitro and in vivo assay revealed that elevated miR-601 inhibited the proliferative, migratory and invasive capacities in OS cells. Mechanically, miR-601 exerted its function via targeting oncogene protein kinase membrane associated tyrosine/threonine 1 (PKMYT1) at post-transcriptional level. Moreover, miR-601 was attenuated by c-Myb at transcriptional level. Taken together, our studies reveal that miR-601 is a suppressive gene negatively correlated with malignancy of OS.


Asunto(s)
Neoplasias Óseas , MicroARNs , Osteosarcoma , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas c-myb , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Biología Computacional , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Osteosarcoma/genética , Osteosarcoma/patología , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo
7.
Bioengineered ; 13(4): 10313-10323, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35436412

RESUMEN

The aim of this study was to investigate whether HUCMSCsWnt10b could promote long bone fracture healing. Commercially-available HUCMSCsEmp (human umbilical cord mesenchymal stem cells transfected with empty vector) in hydrogel, HUCMSCsWnt10b in hydrogel and HUCMSCsWnt10b with the Wnt signaling pathway inhibitor IWR-1 were transplanted into the fracture site in a rat model of femoral fracture. We found that transplantation of HUCMSCsWnt10b significantly accelerated bone healing in a rat model of femoral fracture. Meanwhile, three-point bending test proved that the mechanical properties of the bone at the fracture site in the HUCMSCWnt10b treatment group were significantly better than those of the other treatment groups. To understand the cellular mechanism, we explored the viability of periosteal stem cells (PSCs), as they contribute the greatest number of osteoblast lineage cells to the callus. In line with in vivo data, we found that conditioned medium from HUCMSCsWnt10b enhanced the migration and osteogenic differentiation of PSCs. Furthermore, conditioned medium from HUCMSCsWnt10b also induced endothelial cells to form capillary-like structures in a tube formation assay, which was blocked by SU5416, an angiogenesis inhibitor, suggesting that enhanced vessel formation and growth also contribute to accelerated hard callus formation. In summary, our study demonstrates that HUCMSCsWnt10b promote fracture healing via accelerated hard callus formation, possibly due to enhanced osteogenic differentiation of PSCs and vessel growth. Therefore, HUCMSCsWnt10b may be a promising treatment for long bone fractures.


Asunto(s)
Fracturas del Fémur , Células Madre Mesenquimatosas , Animales , Remodelación Ósea , Cartílago , Diferenciación Celular , Medios de Cultivo Condicionados/metabolismo , Células Endoteliales , Fracturas del Fémur/metabolismo , Fracturas del Fémur/terapia , Curación de Fractura , Hidrogeles , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Ratas , Cordón Umbilical , Vía de Señalización Wnt
8.
J Exp Clin Cancer Res ; 40(1): 294, 2021 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34544449

RESUMEN

BACKGROUND: Long non-coding RNA (LncRNA) controls cell proliferation and plays a significant role in the initiation and progression of esophageal squamous cell carcinoma (ESCC). N6-methyladenosine (m6A) modification now is recognized as a master driver of RNA function to maintain homeostasis in cancer cells. However, how m6A regulates LncRNA function and its role in tumorigenesis of ESCC remain unclear. METHODS: Multiple ESCC datasets were used to analyze gene expression in tumor tissues and normal tissues. Kaplan-Meier method and the ROC curve were conducted to evaluate the prognostic value and diagnostic value of LINC00022 in ESCC, respectively. Both gain-of-function and loss-of-function experiments were employed to investigate the effects of LINC00022 on ESCC growth in vitro and in vivo. Bioinformatics analysis, colorimetric m6A assay, RIP, MeRIP and co-IP was performed to explore the epigenetic mechanism of LINC00022 up-regulation in ESCC. RESULTS: Here we report that m6A demethylation of LncRNA LINC00022 by fat mass and obesity-associated protein (FTO) promotes tumor growth of ESCC in vivo. Clinically, we revealed that LINC00022 was up-regulated in primary ESCC samples and was predictive of poor clinical outcome for ESCC patients. Mechanistically, LINC00022 directly binds to p21 protein and promotes its ubiquitination-mediated degradation, thereby facilitating cell-cycle progression and proliferation. Further, the elevated FTO in ESCC decreased m6A methylation of LINC00022 transcript, leading to the inhibition of LINC00022 decay via the m6A reader YTHDF2. Over-expression of FTO was shown to drive LINC00022-dependent cell proliferation and tumor growth of ESCC. CONCLUSIONS: Thus, this study demonstrated m6A-mediated epigenetic modification of LncRNA contributes to the tumorigenesis in ESCC and LINC00022, specific target of m6A, serves as a potential biomarker for this malignancy.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , ARN Mensajero/genética , Animales , Línea Celular Tumoral , Metilación de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Carcinoma de Células Escamosas de Esófago/mortalidad , Carcinoma de Células Escamosas de Esófago/patología , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Masculino , Ratones , ARN Mensajero/metabolismo
9.
J Orthop Translat ; 23: 21-28, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32455113

RESUMEN

BACKGROUND/OBJECTIVES: Articular cartilage erosion probably plays a substantial role in osteoarthritis (OA) initiation and development. Studies demonstrated that umbilical cord-derived mesenchymal stem cells (UCMSCs) could delay chondrocytes apoptosis and ameliorate OA progression in patients, but the detailed mechanisms are largely uncharacterised. In this study, we aimed to study the effects of UCMSCs on monosodium iodoacetate (MIA)-induced rat OA model, and explore the cellular mechanism of this effect. METHODS: Intra-articular injection of 0.3 â€‹mg MIA in 50 â€‹µL saline was performed on the left knee of the 200 â€‹g weight male Sprague-Dawley rat to induce rat knee OA. A single dose of 2.5 â€‹× â€‹105 undifferentiated UCMSCs one day after MIA or three-time intra-articular injection of 2.5 â€‹× â€‹105 UCMSCs on Days 1, 7 â€‹and 14 were given, respectively. Four weeks after MIA, joints were harvested and processed for paraffin sections. Safranine-O staining, haematoxylin and eosin staining â€‹and immunohistochemistry of MMP-13, ADAMTS-5, Col-2, CD68 â€‹and CD4 were performed to observe cartilage erosion and synovium. For in vitro â€‹studies, migration ability of cartilage superficial layer cells (SFCs) by UCMSCs were accessed by transwell assay. Furthermore, catabolism change of MIA-induced SFCs by UCMSCs was performed by real-rime polymerase chain reaction of Col-X and BCL-2 genes. CCK-8 assay was performed to check proliferation ability of SFCs by UCMSCs-conditioned media. RESULT: In this study, we locally injected human UCMSCs, which is highly proliferative and noninvasively collectible, into MIA-induced rat knee OA. An important finding is on obviously ameliorated cartilage erosion and decreased OA Mankin score by repeated UCMSCs injection after MIA injection compared with single injection, both of which attenuated OA progression compared with vehicle. Interestingly, we observed significantly increased number of SFCs on the articular cartilage surface, probably related to elevated proliferation, mobilisation and inhibited catabolism marker: Col-X and BCL-2 gene expression of cultured SFCs by UCMSCs-conditioned media treatment in vitro. In addition to the change of unique SFCs, catabolism markers of ADAMTS-5 and MMP-13 were substantially upregulated in the whole cartilage layer chondrocytes as well. Strikingly, MIA-induced inflammatory cells infiltration, on both CD4+ Th cells and CD68+ macrophages, and hyperplasia of the synovium, which was alleviated by repeated UCMSCs injection. CONCLUSION: Our study demonstrated a critical role of repeated UCMSCs dosing on preserving SFCs function, cartilage structure and inhibiting synovitis during OA progression, and thus provided mechanistic proof of evidence for the use of UCMSCs on OA patients in the future. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: UCMSCs are a relatively "young" stem cell, and noninvasively collectible. In our study, we clearly demonstrated that it could effectively delay OA progression, possibly through reserving SFCs function and inhibiting synovitis. Therefore, it could be a new promising therapeutic cell source for OA after further clinical trials.

10.
J Orthop Translat ; 23: 29-37, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32477867

RESUMEN

BACKGROUND/OBJECTIVES: Accelerating the process of bone regeneration is of great interest for surgeons and basic scientists alike. Recently, umbilical cord mesenchymal stem cells (UCMSCs) are considered clinically applicable for tissue regeneration due to their noninvasive harvesting and better viability. Nonetheless, the bone regenerative ability of human UCMSCs (HUCMSCs) is largely unknown. This study aimed to investigate whether Wnt10b-overexpressing HUCMSCs have enhanced bone regeneration ability in a rat model. METHOD: A rat calvarial defect was performed on 8-week old male Sprague Dawley rats. Commercially purchased HUCMSCsEmp in hydrogel, HUCMSCsWnt10b in hydrogel and HUCMSCsWnt10b with IWR-1 were placed in the calvarial bone defect right after surgery on rats (N = 8 rats for each group). Calvaria were harvested for micro-CT analysis and histology four weeks after surgery. CFU-F and multi-differentiation assay by oil red staining, alizarin red staining and RT-PCR (real-time polymerase chain reaction) were performed on HUCMSCsEmp and HUCMSCsWnt10b in vitro. Conditioned media from HUCMSCsEmp and HUCMSCsWnt10b were collected and used to treat human umbilical cord vein endothelial cells in Matrigel to access vessel formation capacity by tube formation assay. RESULTS: Alizarin red staining, oil red staining and RT-PCR results showed robust osteogenic differentiation but poor adipogenic differentiation ability of HUCMSCsWnt10b. Furthermore, HUCMSCsWnt10b could accelerate bone defect healing, which was likely due to enhanced angiogenesis after the HUCMSCsWnt10b treatment, because more CD31+ vessels and increased vascular endothelial growth factor-A (VEGF-A) expression were observed, compared with the HUCMSCsEmp treatment. Conditioned media from HUCMSCsWnt10b also induced endothelial cells to form vessel tubes in a tube formation assay, which could be abolished by SU5416, an angiogenesis inhibitor. CONCLUSION: To our knowledge, this is the first study providing empirical evidence that HUCMSCsWnt10b can enhance their ability to heal calvarial bone defects via VEGF-mediated angiogenesis. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: HUCMSCsWnt10b can accelerate critical size calvaria and are a new promising therapeutic cell source for fracture nonunion healing.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA