Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.063
Filtrar
1.
ACS Appl Mater Interfaces ; 16(34): 44623-44635, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39145889

RESUMEN

Injectable extracellular matrix (iECM) is a versatile biological material with beneficial properties such as good degradability, promotion of cell survival, immunomodulation, and facilitation of vascular formation. However, intravenous injection of iECM faces challenges like a short retention time in vivo and low concentration at the lesion site. To address these issues, we prepared a composite hydrogel composed of sodium alginate and iECM and administered it via intrapericardial injection, forming a structure akin to cardiac patches within the pericardium. Compared with intramyocardial injection, intrapericardial injection avoids direct myocardial injury and ectopic tumor formation, offering less invasiveness and better biocompatibility. This study demonstrates that the sodium alginate/infusible extracellular matrix (SA/iECM) composite hydrogel can effectively prolong the local retention time of iECM in the heart, enhance electrical conduction between cardiomyocytes, promote angiogenesis at ischemic myocardial sites, inhibit apoptosis in the infarcted region, mitigate left ventricular remodeling postmyocardial infarction (MI), and improve cardiac function after infarction. Precise coordination of cardiomyocyte contraction and relaxation depends on the rhythmic occurrence of calcium-dependent action potentials. Cardiac dysfunction is partially attributed to the disruption of the excitation-contraction coupling (ECC) mechanism, which is associated with prolonged intracellular Ca2+ transients and alterations in contraction and relaxation Ca2+ levels. Our results show that the SA/iECM composite hydrogel improves electrical conduction, as evidenced by increased Cx43 expression and enhanced intercellular electrical connectivity. This research establishes that intrapericardial injection of a SA/iECM composite hydrogel is a safe and effective treatment modality, providing a theoretical basis for the use of biomaterials in MI therapy.


Asunto(s)
Alginatos , Matriz Extracelular , Hidrogeles , Infarto del Miocardio , Neovascularización Fisiológica , Pericardio , Alginatos/química , Alginatos/farmacología , Animales , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Hidrogeles/química , Hidrogeles/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Pericardio/efectos de los fármacos , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Masculino , Ratas , Angiogénesis
2.
Biosens Bioelectron ; 264: 116668, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39173340

RESUMEN

Traditional hepatocellular carcinoma-chip models lack the cell structure and microenvironments necessary for high pathophysiological correlation, leading to low accuracy in predicting drug efficacy and high production costs. This study proposed a decellularized hepatocellular carcinoma-on-a-chip model to screen anti-tumor nanomedicine. In this model, human hepatocellular carcinoma (HepG2) and human normal liver cells (L02) were co-cultured on a three-dimensional (3D) decellularized extracellular matrix (dECM) in vitro to mimic the tumor microenvironments of human hepatocellular carcinoma in vivo. Additionally, a smart nanomedicine was developed by encapsulating doxorubicin (DOX) into the ferric oxide (Fe3O4)-incorporated liposome nanovesicle (NLV/Fe+DOX). NLV/Fe+DOX selectively killed 78.59% ± 6.78% of HepG2 cells through targeted delivery and synergistic chemo-chemodynamic-photothermal therapies, while the viability of surrounding L02 cells on the chip model retained high, at over 90.0%. The drug efficacy tested using this unique chip model correlated well with the results of cellular and animal experiments. In summary, our proposed hepatocellular carcinoma-chip model is a low-cost yet accurate drug-testing platform with significant potential for drug screening.


Asunto(s)
Carcinoma Hepatocelular , Doxorrubicina , Dispositivos Laboratorio en un Chip , Neoplasias Hepáticas , Nanomedicina , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Doxorrubicina/farmacología , Doxorrubicina/química , Doxorrubicina/uso terapéutico , Células Hep G2 , Nanomedicina/métodos , Animales , Liposomas/química , Matriz Extracelular/química , Matriz Extracelular/efectos de los fármacos , Compuestos Férricos/química , Técnicas Biosensibles/métodos , Microambiente Tumoral/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico
3.
Int Immunopharmacol ; 140: 112887, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39116493

RESUMEN

Osteoarthritis (OA) is a prevalent disease of the musculoskeletal system that causes functional deterioration and diminished quality of life. Myrislignan (MRL) has a wide range of pharmacological characteristics, including an anti-inflammatory ability. Although inflammation is a major cause of OA, the role of MRL in OA treatment is still not well-understood. In this study, we analyze the anti-inflammatory and anti-ECM degradation effects of MRL both in vivo and in vitro. Rat primary chondrocytes were treated with interleukin-1ß (IL-1ß) to simulate inflammatory environmental conditions and OA in vitro. The in vivo OA rat model was established by anterior cruciate ligament transection (ACLT) on rat. Our investigation discovered that MRL lowers the IL-1ß-activated tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX2) and inducible nitric-oxide synthase (iNOS) expression in chondrocytes. Moreover, MRL effectively alleviates IL-1ß-induced extracellular matrix (ECM) degradation and promotes ECM synthesis in chondrocytes by upregulating the mRNA level expression of collagen-II and aggrecan (ACAN), downregulating the expression of matrix metalloproteinases-3,-13 (MMP-3, MMP-13), and a disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5). Gene expression profiles of different groups identified DEGs that were mainly enriched in functions associated with NF-κB signaling pathway, and other highly enriched in functions related to TNF, IL-17, Rheumatoid arthritis and cytokine-cytokine receptor signaling pathways. Venn interaction of DEGs from the abovementioned five pathways showed that Nfkbia, Il1b, Il6, Nfkb1, Ccl2, Mmp3 were highly enriched DEGs. In addition, our research revealed that MRL suppresses NF-κB and modulates the Nrf2/HO-1/JNK signaling pathway activated by IL-1ß in chondrocytes. In vivo research shows that MRL slows the progression of OA in rats. Our findings imply that MRL might be a viable OA therapeutic choice.


Asunto(s)
Condrocitos , Interleucina-1beta , Lignanos , Osteoartritis , Ratas Sprague-Dawley , Animales , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Células Cultivadas , Interleucina-1beta/metabolismo , Masculino , Ratas , Lignanos/farmacología , Lignanos/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , FN-kappa B/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Progresión de la Enfermedad , Factor de Necrosis Tumoral alfa/metabolismo , Ciclooxigenasa 2/metabolismo , Ciclooxigenasa 2/genética , Modelos Animales de Enfermedad , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Transducción de Señal/efectos de los fármacos , Proteína ADAMTS5/metabolismo , Proteína ADAMTS5/genética , Humanos
4.
Int Immunopharmacol ; 139: 112717, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39067404

RESUMEN

Intervertebral disc degeneration (IVDD), a common degenerative disc disease, is a major etiological factor for back pain, affecting a significant number of middle-aged and elderly individuals worldwide. Thus, IVDD is a major socio-economic burden. The factors contributing to the complex IVDD etiology, which has not been elucidated, include inflammation, oxidative stress, and natural aging. In particular, inflammation and aging of nucleus pulposus cells are considered primary pathogenic factors. Isorhapontigenin (ISO) is a polyphenolic compound commonly found in traditional Chinese herbs and grapes. We have demonstrated that ISO exerts anti-inflammatory and anti-aging effects and mitigates extracellular matrix (ECM) degradation. In this study, in vitro experiments revealed that, ISO delays aging and ECM degradation by promoting PI3K/AKT/mTOR-mediated autophagy. Meanwhile, in vivo experiments affirmed that ISO delays the progression of IVDD.


Asunto(s)
Autofagia , Senescencia Celular , Matriz Extracelular , Degeneración del Disco Intervertebral , Núcleo Pulposo , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Degeneración del Disco Intervertebral/tratamiento farmacológico , Degeneración del Disco Intervertebral/metabolismo , Núcleo Pulposo/efectos de los fármacos , Núcleo Pulposo/metabolismo , Núcleo Pulposo/patología , Serina-Treonina Quinasas TOR/metabolismo , Autofagia/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Transducción de Señal/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Senescencia Celular/efectos de los fármacos , Masculino , Ratas Sprague-Dawley , Humanos , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Ratas , Células Cultivadas , Estilbenos
5.
Biomater Adv ; 163: 213936, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38959652

RESUMEN

Matrix stiffening is one of the major risk factors for hepatocellular carcinoma (HCC) and drives tumor progression. The extracellular matrix (ECM) stiffness of HCC displays mechanical heterogeneity, with stiffness increasing from the core to the invasive frontier. The distribution of liver cancer stem cells (CSCs) is related to this mechanical property. However, it is not sufficiently understood how heterogeneous matrix stiffness regulates the stemness of CSCs. In this study, we developed an adjustable gelatin/alginate hydrogel to investigate the effect of various matrix stiffnesses on CSC stemness under three-dimensional culture conditions. Gelatin/alginate hydrogel with the stiffness of soft (5 kPa), medium (16 kPa), and stiff (81 kPa) were prepared by altering the concentration of calcium ions. It was found that a stiffer matrix promoted stemness-associated gene expression, reduced drug sensitivity, enhanced sphere-forming and clonogenic ability, and tumorigenic potential. Mechanistically, matrix stiffening facilitates CSC stemness by increasing Yes-associated protein (YAP) activity and inhibiting Bcl-2 modifying factor (BMF) expression. Knockdown of YAP or overexpression of BMF significantly attenuated matrix stiffening-induced stemness, suggesting the involvement of YAP and BMF in this process. Together, our results unravel the regulatory mechanism of heterogeneous matrix stiffness on CSC stemness and also provide a novel therapeutic strategy for eradicating CSCs and improving the efficiency of HCC treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Carcinoma Hepatocelular , Matriz Extracelular , Hidrogeles , Neoplasias Hepáticas , Células Madre Neoplásicas , Factores de Transcripción , Proteínas Señalizadoras YAP , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/metabolismo , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Hidrogeles/química , Proteínas Señalizadoras YAP/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Alginatos/farmacología , Animales , Gelatina/química , Ratones
6.
ACS Appl Mater Interfaces ; 16(31): 40555-40569, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39042857

RESUMEN

Regenerating periodontal defects in osteoporosis patients presents a significant clinical challenge. Unlike the relatively straightforward regeneration of homogeneous bone tissue, periodontal regeneration requires the intricate reconstruction of the cementum-periodontal ligament-alveolar bone interface. Strontium (Sr)-doped biomaterials have been extensively utilized in bone tissue engineering due to their remarkable pro-osteogenic attributes. However, their application in periodontal tissue regeneration has been scarcely explored. In this study, we synthesized an innovative injectable Sr-BGN/GNM scaffold by integrating Sr-doped bioactive glass nanospheres (Sr-BGNs) into the nanofiber architecture of gelatin nanofiber microspheres (GNMs). This design, mimicking the natural bone extracellular matrix (ECM), enhanced the scaffold's mechanical properties and effectively controlled the sustained release of Sr ions (Sr2+), thereby promoting the proliferation, osteogenic differentiation, and ECM secretion of PDLSCs and BMSCs, as well as enhancing vascularization in endothelial cells. In vivo experiments further indicated that the Sr-BGNs/GNMs significantly promoted osteogenesis and angiogenesis. Moreover, the scaffold's tunable degradation kinetics optimized the prolonged release and pro-regenerative effects of Sr2+ in vivo, matching the pace of periodontal regeneration and thereby facilitating the regeneration of functional periodontal tissues under osteoporotic conditions. Therefore, Sr-BGNs/GNMs emerge as a promising candidate for advancing periodontal regeneration strategies.


Asunto(s)
Matriz Extracelular , Microesferas , Nanofibras , Osteoporosis , Estroncio , Estroncio/química , Estroncio/farmacología , Nanofibras/química , Osteoporosis/tratamiento farmacológico , Humanos , Matriz Extracelular/química , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Animales , Osteogénesis/efectos de los fármacos , Andamios del Tejido/química , Diferenciación Celular/efectos de los fármacos , Ingeniería de Tejidos , Proliferación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Regeneración/efectos de los fármacos
7.
ACS Biomater Sci Eng ; 10(8): 4682-4700, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-38967485

RESUMEN

Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.


Asunto(s)
Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Matriz Extracelular , Mecanotransducción Celular , Neoplasias , Humanos , Resistencia a Antineoplásicos/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/metabolismo , Mecanotransducción Celular/efectos de los fármacos , Autofagia/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología
8.
ACS Biomater Sci Eng ; 10(8): 5168-5180, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39016069

RESUMEN

The radiation-induced skin injury (RISI) remains a great challenge for clinical wound management and care after radiotherapy, as patients will suffer from the acute radiation injury and long-term chronic inflammatory damage during the treatment. The excessive ROS in the early acute stage and prolonged inflammatory response in the late healing process always hinder therapeutic efficiency. Herein, we developed an extracellular matrix (ECM)-mimetic multifunctional glycopeptide hydrogel (oCP@As) to promote and accelerate RISI repair via a dual-modulation strategy in different healing stages. The oCP@As hydrogel not only can form an ECM-like nanofiber structure through the Schiff base reaction but also exhibits ROS scavenging and DNA double-strand break repair abilities, which can effectively reduce the acute radiation damage. Meanwhile, the introduction of oxidized chondroitin sulfate, which is the ECM polysaccharide-like component, enables regulation of the inflammatory response by adsorption of inflammatory factors, accelerating the repair of chronic inflammatory injury. The animal experiments demonstrated that oCP@As can significantly weaken RISI symptoms, promote epidermal tissue regeneration and angiogenesis, and reduce pro-inflammatory cytokine expression. Therefore, this multifunctional glycopeptide hydrogel dressing can effectively attenuate RISI symptoms and promote RISI healing, showing great potential for clinical applications in radiotherapy protection and repair.


Asunto(s)
Glicopéptidos , Hidrogeles , Traumatismos por Radiación , Piel , Cicatrización de Heridas , Animales , Hidrogeles/química , Hidrogeles/farmacología , Glicopéptidos/química , Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Piel/efectos de los fármacos , Piel/efectos de la radiación , Piel/patología , Traumatismos por Radiación/tratamiento farmacológico , Traumatismos por Radiación/prevención & control , Cicatrización de Heridas/efectos de los fármacos , Ratones , Humanos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Masculino
9.
Neurochem Res ; 49(10): 2743-2762, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38951281

RESUMEN

The purpose of this study is to explore the shared molecular pathogenesis of traumatic brain injury (TBI) and high-grade glioma and investigate the mechanism of propofol (PF) as a potential protective agent. By analyzing the Chinese glioma genome atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases, we compared the transcriptomic data of high-grade glioma and TBI patients to identify common pathological mechanisms. Through bioinformatics analysis, in vitro experiments and in vivo TBI model, we investigated the regulatory effect of PF on extracellular matrix (ECM)-related genes through Prrx1 under oxidative stress. The impact of PF on BBB integrity under oxidative stress was investigated using a dual-layer BBB model, and we explored the protective effect of PF on tight junction proteins and ECM-related genes in mice after TBI. The study found that high-grade glioma and TBI share ECM instability as an important molecular pathological mechanism. PF stabilizes the ECM and protects the BBB by directly binding to Prrx1 or indirectly regulating Prrx1 through miRNAs. In addition, PF reduces intracellular calcium ions and ROS levels under oxidative stress, thereby preserving BBB integrity. In a TBI mouse model, PF protected BBB integrity through up-regulated tight junction proteins and stabilized the expression of ECM-related genes. Our study reveals the shared molecular pathogenesis between TBI and glioblastoma and demonstrate the potential of PF as a protective agent of BBB. This provides new targets and approaches for the development of novel neurotrauma therapeutic drugs.


Asunto(s)
Barrera Hematoencefálica , Lesiones Traumáticas del Encéfalo , Matriz Extracelular , Glioma , Propofol , Propofol/farmacología , Propofol/uso terapéutico , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Glioma/metabolismo , Glioma/tratamiento farmacológico , Glioma/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Ratones , Humanos , Masculino , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Estrés Oxidativo/efectos de los fármacos , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Línea Celular Tumoral
10.
Discov Med ; 36(186): 1441-1452, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39054715

RESUMEN

BACKGROUND: Synovial inflammation plays a crucial role in osteoarthritis (OA). Gastrodin (GAS), an active ingredient derived from the Gastrodia elata Blume rhizome, possesses antioxidant and anti-inflammatory pharmacological effects. This research aimed to evaluate the function and molecular mechanism of GAS on human fibroblast-like synoviocytes of osteoarthritis (HFLS-OA) induced by interleukin (IL)-1ß. METHODS: The impact of GAS on the viability of IL-1ß-treated HFLS-OA cells was assessed using the cell counting kit-8 (CCK-8). Quantitative real-time reverse transcription PCR (qRT-PCR) was employed to detect changes in IL-8, IL-6, monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor (TNF)-α, and Gremlin-1 mRNA expression in each group. Corresponding kits were utilized to measure the catalase (CAT) and superoxide dismutase (SOD) activities, as well as the nitric oxide (NO) level. Western blot analysis was conducted to examine the expression of extracellular matrix degradation-associated proteins and nuclear factor kappa-B (NF-κB) pathway-correlated proteins in each group. RESULTS: GAS significantly promoted the proliferation of IL-1ß-induced HFLS-OA cells and concurrently down-regulated Gremlin-1 mRNA expression (p < 0.05). Through the down-regulation of Gremlin-1 expression, GAS exhibited the following effects: decreased IL-8, IL-6, and TNF-α mRNA expression, as well as NO levels (p < 0.05); increased SOD and CAT activities (p < 0.05); down-regulated matrix metallopeptidase 13 (MMP-13) and MMP-1 protein expression levels (p < 0.01); and up-regulated collagen II protein expression level (p < 0.01) in IL-1ß-treated HFLS-OA cells. Additionally, GAS decreased phospho-inhibitory kappa B (p-IκB)/IκB, phospho-inhibitory kappa B kinase (p-IKK)/IKK, and p-p65/p65 ratios in IL-1ß-induced HFLS-OA cells by inhibiting Gremlin-1 expression (p < 0.01). CONCLUSION: GAS demonstrates a positive impact on inflammation, oxidative stress, and extracellular matrix degradation in IL-1ß-mediated HFLS-OA cells. This effect is achieved by suppressing Gremlin-1 expression and reducing NF-κB pathway activity.


Asunto(s)
Alcoholes Bencílicos , Matriz Extracelular , Glucósidos , Inflamación , Interleucina-1beta , FN-kappa B , Estrés Oxidativo , Sinoviocitos , Humanos , Glucósidos/farmacología , Interleucina-1beta/metabolismo , Alcoholes Bencílicos/farmacología , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inflamación/metabolismo , Inflamación/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Sinoviocitos/efectos de los fármacos , Sinoviocitos/metabolismo , Sinoviocitos/patología , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Osteoartritis/patología , Osteoartritis/metabolismo , Osteoartritis/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intercelular
11.
Acta Neuropathol Commun ; 12(1): 102, 2024 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-38907342

RESUMEN

Neurofibromatosis Type 1 (NF1) is caused by loss of function variants in the NF1 gene. Most patients with NF1 develop skin lesions called cutaneous neurofibromas (cNFs). Currently the only approved therapeutic for NF1 is selumetinib, a mitogen -activated protein kinase (MEK) inhibitor. The purpose of this study was to analyze the transcriptome of cNF tumors before and on selumetinib treatment to understand both tumor composition and response. We obtained biopsy sets of tumors both pre- and on- selumetinib treatment from the same individuals and were able to collect sets from four separate individuals. We sequenced mRNA from 5844 nuclei and identified 30,442 genes in the untreated group and sequenced 5701 nuclei and identified 30,127 genes in the selumetinib treated group. We identified and quantified distinct populations of cells (Schwann cells, fibroblasts, pericytes, myeloid cells, melanocytes, keratinocytes, and two populations of endothelial cells). While we anticipated that cell proportions might change with treatment, we did not identify any one cell population that changed significantly, likely due to an inherent level of variability between tumors. We also evaluated differential gene expression based on drug treatment in each cell type. Ingenuity pathway analysis (IPA) was also used to identify pathways that differ on treatment. As anticipated, we identified a significant decrease in ERK/MAPK signaling in cells including Schwann cells but most specifically in myeloid cells. Interestingly, there is a significant decrease in opioid signaling in myeloid and endothelial cells; this downward trend is also observed in Schwann cells and fibroblasts. Cell communication was assessed by RNA velocity, Scriabin, and CellChat analyses which indicated that Schwann cells and fibroblasts have dramatically altered cell states defined by specific gene expression signatures following treatment (RNA velocity). There are dramatic changes in receptor-ligand pairs following treatment (Scriabin), and robust intercellular signaling between virtually all cell types associated with extracellular matrix (ECM) pathways (Collagen, Laminin, Fibronectin, and Nectin) is downregulated after treatment. These response specific gene signatures and interaction pathways could provide clues for understanding treatment outcomes or inform future therapies.


Asunto(s)
Bencimidazoles , Matriz Extracelular , Células de Schwann , Transducción de Señal , Neoplasias Cutáneas , Humanos , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Células de Schwann/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Bencimidazoles/farmacología , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Transducción de Señal/efectos de los fármacos , Neurofibroma/genética , Neurofibroma/tratamiento farmacológico , Neurofibroma/metabolismo , Neurofibroma/patología , Femenino , Masculino , RNA-Seq , Persona de Mediana Edad , Adulto , Neurofibromatosis 1/genética , Neurofibromatosis 1/tratamiento farmacológico , Neurofibromatosis 1/patología , Inhibidores de Proteínas Quinasas/farmacología , Transcriptoma/efectos de los fármacos
12.
ACS Appl Mater Interfaces ; 16(24): 30929-30957, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38832934

RESUMEN

Bioengineered composite hydrogel platforms made of a supramolecular coassembly have recently garnered significant attention as promising biomaterial-based healthcare therapeutics. The mechanical durability of amyloids, in conjunction with the structured charged framework rendered by biologically abundant key ECM component glycosaminoglycan, enables us to design minimalistic customized biomaterial suited for stimuli responsive therapy. In this study, by harnessing the heparin sulfate-binding aptitude of amyloid fibrils, we have constructed a pH-responsive extracellular matrix (ECM) mimicking hydrogel matrix. This effective biocompatible platform comprising heparin sulfate-amyloid coassembled hydrogel embedded with polyphenol functionalized silver nanoparticles not only provide a native skin ECM-like conductive environment but also provide wound-microenvironment responsive on-demand superior antibacterial efficacy for effective diabetic wound healing. Interestingly, both the cytocompatibility and antibacterial properties of this bioinspired matrix can be fine-tuned by controlling the mutual ratio of heparin sulfate-amyloid and incubated silver nanoparticle components, respectively. The designed biomaterial platform exhibits notable effectiveness in the treatment of chronic hyperglycemic wounds infected with multidrug-resistant bacteria, because of the integration of pH-responsive release characteristics of the incubated functionalized AgNP and the antibacterial amyloid fibrils. In addition to this, the aforementioned assemblage shows exceptional hemocompatibility with significant antibiofilm and antioxidant characteristics. Histological evidence of the incised skin tissue sections indicates that the fabricated composite hydrogel is also effective in controlling pro-inflammatory cytokines such as IL6 and TNFα expressions at the wound vicinity with significant upregulation of angiogenesis markers like CD31 and α-SMA.


Asunto(s)
Amiloide , Antibacterianos , Matriz Extracelular , Heparina , Hidrogeles , Nanopartículas del Metal , Plata , Cicatrización de Heridas , Cicatrización de Heridas/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Heparina/química , Heparina/farmacología , Plata/química , Plata/farmacología , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Nanopartículas del Metal/química , Amiloide/química , Amiloide/metabolismo , Animales , Humanos , Staphylococcus aureus/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología
13.
Sci Rep ; 14(1): 14178, 2024 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898058

RESUMEN

Increasing evidence supports the hypothesis that cancer progression is under mitochondrial control. Mitochondrial fission plays a pivotal role in the maintenance of cancer cell homeostasis. The inhibition of DRP1, the main regulator of mitochondrial fission, with the mitochondrial division inhibitor (mdivi-1) had been associated with cancer cell sensitivity to chemotherapeutics and decrease proliferation. Here, using breast cancer cells we find that mdivi-1 induces the detachment of the cells, leading to a bulk of floating cells that conserved their viability. Despite a decrease in their proliferative and clonogenic capabilities, these floating cells maintain the capacity to re-adhere upon re-seeding and retain their migratory and invasive potential. Interestingly, the cell detachment induced by mdivi-1 is independent of DRP1 but relies on inhibition of mitochondrial complex I. Furthermore, mdivi-1 induces cell detachment rely on glucose and the pentose phosphate pathway. Our data evidence a novel DRP1-independent effect of mdivi-1 in the attachment of cancer cells. The generation of floating viable cells restricts the use of mdivi-1 as a therapeutic agent and demonstrates that mdivi-1 effect on cancer cells are more complex than anticipated.


Asunto(s)
Neoplasias de la Mama , Dinaminas , Matriz Extracelular , Dinámicas Mitocondriales , Quinazolinonas , Humanos , Dinaminas/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Línea Celular Tumoral , Quinazolinonas/farmacología , Dinámicas Mitocondriales/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos
14.
Phytother Res ; 38(7): 3509-3524, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38695125

RESUMEN

Osteoarthritis (OA), a joint disease that is associated with inflammatory processes is involved in joint destruction. Scutellarein (Scu), a component of the medicinal herbs Scutellaria barbata D. Don and Erigeron breviscapus (vant) Hand Mass, has anti-inflammatory effects. We explored the role of Scu in the development of OA and the underlying mechanisms. CCK-8 assays, Calcein-AM/PI and EdU staining were used to determine chondrocyte viability after Scu exposure. Western blot, qPCR, as well as ELISA were utilized to measure extracellular matrix (ECM) degradation and inflammation. Immunofluorescence (IF), western blot and luciferase assays were used to examine the NF-kappaB (NF-κB) pathway. Scu interacting proteins were predicted using network pharmacology analysis and molecular docking. X-ray, H&E, Safranin O-Fast Green(S-O), toluidine blue, and immunohistochemistry analysis were used to examine the therapeutic effects of Scu in OA using destabilization of medial meniscus (DMM) models. Scu demonstrated inhibitory effects on ECM degradation and pro-inflammatory factor levels in chondrocytes treated with IL-1ß. Mechanistically, Scu inhibited the IL-1ß-induced activation of the PI3K/Akt/ NF-κB signaling pathway cascades. Furthermore, Scu has been shown to have significant binding capacities to PI3K. Additionally, Scu ameliorated the OA progression in DMM models. Our findings suggest that Scu may contribute to the amelioration of OA progression by targeting the PI3K/Akt/NF-κB signaling pathway, implying Scu possesses promising therapeutic potential for the treatment of OA.


Asunto(s)
Apigenina , Condrocitos , FN-kappa B , Osteoartritis , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Osteoartritis/tratamiento farmacológico , FN-kappa B/metabolismo , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Apigenina/farmacología , Masculino , Simulación del Acoplamiento Molecular , Ratas Sprague-Dawley , Antiinflamatorios/farmacología , Ratas , Humanos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos
15.
J Pharm Pharmacol ; 76(8): 1018-1027, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-38776436

RESUMEN

OBJECTIVES: This study aimed to reveal the anti-fibrotic effects of Botrychium ternatum (Thunb.) Sw. (BT) against idiopathic pulmonary fibrosis (IPF) and to preliminarily analyze its potential mechanism on bleomycin-induced IPF rats. METHODS: The inhibition of fibrosis progression in vivo was assessed by histopathology combined with biochemical indicators. In addition, the metabolic regulatory mechanism was investigated using 1H-nuclear magnetic resonance-based metabolomics combined with multivariate statistical analysis. KEY FINDINGS: Firstly, biochemical analysis revealed that BT notably suppressed the expression of hydroxyproline and transforming growth factor-ß1 in the pulmonary tissue. Secondly, Masson's trichrome staining and hematoxylin and eosin showed that BT substantially improved the structure of the damaged lung and significantly inhibited the proliferation of collagen fibers and the deposition of extracellular matrix. Finally, serum metabolomic analysis suggested that BT may exert anti-fibrotic effects by synergistically regulating tyrosine metabolism; phenylalanine, tyrosine and tryptophan biosynthesis; and synthesis and degradation of ketone bodies. CONCLUSIONS: Our study not only clarifies the potential anti-fibrotic mechanism of BT against IPF at the metabolic level but also provides a theoretical basis for developing BT as an effective anti-fibrotic agent.


Asunto(s)
Bleomicina , Fibrosis Pulmonar Idiopática , Pulmón , Metabolómica , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1 , Animales , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/prevención & control , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Metabolómica/métodos , Masculino , Ratas , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Factor de Crecimiento Transformador beta1/metabolismo , Hidroxiprolina/metabolismo , Modelos Animales de Enfermedad , Espectroscopía de Protones por Resonancia Magnética/métodos , Antifibróticos/farmacología , Tirosina/análogos & derivados , Tirosina/metabolismo , Cuerpos Cetónicos/metabolismo , Colágeno/metabolismo , Fenilalanina/farmacología , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Extractos Vegetales/farmacología , Triptófano/metabolismo , Triptófano/farmacología , Medicamentos Herbarios Chinos/farmacología
16.
J Orthop Surg Res ; 19(1): 308, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38773639

RESUMEN

BACKGROUND: Intervertebral disc degeneration (IDD) is an increasingly important cause of low back pain (LBP) that results in substantial health and economic burdens. Inflammatory pathway activation and the production of reactive oxygen species (ROS) play vital roles in the progression of IDD. Several studies have suggested that phillyrin has a protective role and inhibits inflammation and the production of ROS. However, the role of phillyrin in IDD has not been confirmed. PURPOSE: The purpose of this study was to investigate the role of phillyrin in IDD and its mechanisms. STUDY DESIGN: To establish IDD models in vivo, ex-vivo, and in vitro to verify the function of phillyrin in IDD. METHOD: The effects of phillyrin on extracellular matrix (ECM) degeneration, inflammation, and oxidation in nucleus pulposus (NP) cells were assessed using immunoblotting and immunofluorescence analysis. Additionally, the impact of phillyrin administration on acupuncture-mediated intervertebral disc degeneration (IDD) in rats was evaluated using various techniques such as MRI, HE staining, S-O staining, and immunohistochemistry (IHC). RESULT: Pretreatment with phillyrin significantly inhibited the IL-1ß-mediated reduction in the degeneration of ECM and apoptosis by alleviating activation of the NF-κB inflammatory pathway and the generation of ROS. In addition, in vivo and ex-vivo experiments verified the protective effect of phillyrin against IDD. CONCLUSION: Phillyrin can attenuate the progression of IDD by reducing ROS production and activating inflammatory pathways.


Asunto(s)
Progresión de la Enfermedad , Degeneración del Disco Intervertebral , FN-kappa B , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Degeneración del Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/patología , Animales , Especies Reactivas de Oxígeno/metabolismo , FN-kappa B/metabolismo , Ratas , Masculino , Núcleo Pulposo/metabolismo , Núcleo Pulposo/efectos de los fármacos , Núcleo Pulposo/patología , Transducción de Señal/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Cultivadas , Humanos , Apoptosis/efectos de los fármacos
17.
Colloids Surf B Biointerfaces ; 240: 113991, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38815311

RESUMEN

Diabetes mellitus is a chronic metabolic disease with prolonged low-grade inflammation and impaired cellular function, leading to poor wound healing. The treatment of diabetic wounds remains challenging due to the complex wound microenvironment. In view of the prominence of fish scales in traditional Chinese medicine and their wide application in modern medicine, we isolated the intercellular components in the scales of sea bass, obtained a natural composite hydrogel, fish scales gel (FSG), and applied it to diabetic chronic wounds. FSG was rich in collagen-like proteins, and possessed low-temperature gelation properties. In vitro, FSG was biocompatible and promoted fibroblast proliferation by approximately 40 %, endothelial cell migration by approximately 20 % and activated the M1 macrophages. In addition, FSG restored the function of fibroblasts and vascular endothelial cells damaged by high glucose. Importantly, FSG normalized the acute inflammatory response to impaired macrophages in a high-glucose microenvironment. Transcriptome analysis implies that this mechanism may involve enhanced cell signaling and cellular communication, improved sensitivity to cytokines, and activation of the TNF signaling pathway. Animal experiments confirmed that FSG significantly improved wound closure by approximately 15 % in diabetic rats, showing similar effects to acute wounds. In conclusion, the regulation of multiple cellular functions by FSG, especially the counterintuitive ability to induce acute inflammation, promoted diabetic wound healing and provides a novel therapeutic strategy for wound repair in diabetic patients.


Asunto(s)
Diabetes Mellitus Experimental , Hidrogeles , Cicatrización de Heridas , Animales , Cicatrización de Heridas/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Ratas , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Escamas de Animales/química , Ratas Sprague-Dawley , Proliferación Celular/efectos de los fármacos , Masculino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Movimiento Celular/efectos de los fármacos , Ratones , Peces
18.
Biochem Biophys Res Commun ; 721: 150108, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38762931

RESUMEN

Drug-tolerant persister (DTP) cells remain following chemotherapy and can cause cancer relapse. However, it is unclear when acquired resistance to chemotherapy emerges. Here, we compared the gene expression profiles of gastric cancer patient-derived cells (GC PDCs) and their respective xenograft tumors with different sensitivities to 5-fluorouracil (5-FU) by using immunodeficient female BALB/c-nu mice. RNA sequencing analysis of 5-FU-treated PDCs demonstrated that DNA replication/cell cycle-related genes were transiently induced in the earlier phase of DTP cell emergence, while extracellular matrix (ECM)-related genes were sustainably upregulated during long-term cell survival in 5-FU-resistant residual tumors. NicheNet analysis, which uncovers cell-cell signal interactions, indicated the transforming growth factor-ß (TGF-ß) pathway as the upstream regulator in response to 5-FU treatment. This induced ECM-related gene expression in the 5-FU-resistant tumor model. In the 5-FU-resistant residual tumors, there was a marked upregulation of cancer cell-derived TGF-ß1 expression and increased phosphorylation of SMAD3, a downstream regulator of the TGF-ß receptor. By contrast, these responses were not observed in a 5-FU-sensitive tumor model. We further found that TGF-ß-related upregulation of ECM genes was preferentially observed in non-responders to chemotherapy with 5-FU and/or oxaliplatin among 22 patient-derived xenograft tumors. These observations suggest that chemotherapy-induced activation of the TGF-ß1/SMAD3/ECM-related gene axis is a potential biomarker for the emergence of drug resistance in GCs.


Asunto(s)
Resistencia a Antineoplásicos , Matriz Extracelular , Fluorouracilo , Regulación Neoplásica de la Expresión Génica , Ratones Endogámicos BALB C , Transducción de Señal , Neoplasias Gástricas , Factor de Crecimiento Transformador beta , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Resistencia a Antineoplásicos/genética , Humanos , Animales , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Femenino , Transducción de Señal/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Ratones , Factor de Crecimiento Transformador beta/metabolismo , Ratones Desnudos , Línea Celular Tumoral , Proteína smad3/metabolismo , Proteína smad3/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Biochem Pharmacol ; 225: 116280, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38735446

RESUMEN

The pivotal role of human endometrial stromal cells (hESCs) in the development of endometriosis lies in their ability to adopt a pro-invasive and proinflammatory profile upon migration to areas outside the uterus. However, the molecular mechanisms involved in these events remain unclear. In this study, we investigated how angiotensin II (Ang II) affects the plasminogen-plasmin system in hESCs, and the mechanisms underlying cell proliferation, migration, matrix degradation, and inflammation. Precursors, receptors, and peptidases involved in angiotensin metabolism increased significantly in Ang II-treated hESCs. The expression and activity of tissue (tPA)- and urokinase (uPA)- type plasminogen activators and the receptor for uPA (uPAR) were induced in the presence of Ang II. The up-regulation of tPA-uPA/uPAR pathway significantly contributes to heightened plasmin production both on the surface of hESCs and in their conditioned media. As a result, the plasmin generation induced by Ang II enhances the degradation of fibrin and matrix proteins, while also boosting hESC viability, proliferation, and migration through the up-regulation of growth factor expression. Notably, Ang II-induced hESC migration was dependent on the generation of active plasmin on cell surface. Ang II regulates oxidative and inflammatory signalling in hESCs primarily via NADPH oxidase and through the up-regulation of proinflammatory cytokines and adhesion molecules. Interestingly, Ang II receptor (AT1R) blockage, decreased plasmin generation, tPA-uPA/uPAR expression and hESC migration. Our results suggest that Ang II/AT1R axis regulates hESC proliferation and migration through tPA-uPA/uPAR pathway activation and plasmin generation. We propose the Ang II/AT1R axis as a potential target for endometriosis treatment.


Asunto(s)
Angiotensina II , Movimiento Celular , Endometrio , Matriz Extracelular , Fibrinolisina , Plasminógeno , Receptor de Angiotensina Tipo 1 , Transducción de Señal , Células del Estroma , Humanos , Femenino , Endometrio/metabolismo , Endometrio/citología , Endometrio/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Fibrinolisina/metabolismo , Células del Estroma/metabolismo , Células del Estroma/efectos de los fármacos , Angiotensina II/farmacología , Transducción de Señal/fisiología , Transducción de Señal/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Plasminógeno/metabolismo , Células Cultivadas , Inflamación/metabolismo
20.
Int Ophthalmol ; 44(1): 229, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38795168

RESUMEN

BACKGROUND: The multifunctional profibrotic cytokine transforming growth factor-beta2 (TGF-ß2) is implicated in the pathophysiology of primary open angle glaucoma. Paeoniflorin (PAE) is a monoterpene glycoside with multiple pharmacological efficacies, such as antioxidant, anti-fibrotic, and anti-inflammatory properties. Studies have demonstrated that paeoniflorin protects human corneal epithelial cells, retinal pigment epithelial cells, and retinal microglia from damage. Here, the biological role of PAE in TGF-ß2-dependent remodeling of the extracellular matrix (ECM) within the trabecular meshwork (TM) microenvironment. METHODS: Primary or transformed (GTM3) human TM (HTM) cells conditioned in serum-free media were incubated with TGF-ß2 (5 ng/mL). PAE (300 µM) was added to serum-starved confluent cultures of HTM cells for 2 h, followed by incubation with TGF-ß2 for 22 h. SB-431542, a TGF-ß receptor inhibitor (10 µM), was used as a positive control. The levels of intracellular ROS were evaluated by CellROX green dye. Western blotting was used to measure the levels of TGF-ß2/Smad2/3 signaling-related molecules. Collagen 1α1, collagen 4α1, and connective tissue growth factor (CTGF) expression was evaluated by RT-qPCR. Immunofluorescence assay was conducted to measure collagen I/IV expression in HTM cells. Phalloidin staining assay was conducted for evaluating F-actin stress fiber formation in the cells. RESULTS: PAE attenuated TGF-ß2-induced oxidative stress and suppressed TGF-ß2-induced Smad2/3 signaling in primary or transformed HTM cells. Additionally, PAE repressed TGF-ß2-induced upregulation of collagen 1α1, collagen 4α1, and CTGF expression and reduced TGF-ß2-mediated collagen I/IV expression and of F-actin stress fiber formation in primary or transformed HTM cells. CONCLUSION: PAE alleviates TGF-ß2-induced ECM deposition and oxidative stress in HTM cells through inactivation of Smad2/3 signaling.


Asunto(s)
Matriz Extracelular , Glucósidos , Monoterpenos , Estrés Oxidativo , Malla Trabecular , Factor de Crecimiento Transformador beta2 , Humanos , Estrés Oxidativo/efectos de los fármacos , Monoterpenos/farmacología , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta2/farmacología , Glucósidos/farmacología , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Malla Trabecular/efectos de los fármacos , Malla Trabecular/metabolismo , Malla Trabecular/patología , Células Cultivadas , Transducción de Señal/efectos de los fármacos , Western Blotting
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA