Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.601
1.
J Orthop Surg Res ; 19(1): 329, 2024 Jun 02.
Article En | MEDLINE | ID: mdl-38825706

BACKGROUND: Fibrosis is a significant pathological feature of chronic skeletal muscle injury, profoundly affecting muscle regeneration. Fibro-adipogenic progenitors (FAPs) have the ability to differentiate into myofibroblasts, acting as a primary source of extracellular matrix (ECM). the process by which FAPs differentiate into myofibroblasts during chronic skeletal muscle injury remains inadequately explored. METHOD: mouse model with sciatic nerve denervated was constructed and miRNA expression profiles between the mouse model and uninjured mouse were analyzed. qRT/PCR and immunofluorescence elucidated the effect of miR-27b-3p on fibrosis in vivo and in vitro. Dual-luciferase reporter identified the target gene of miR-27b-3p, and finally knocked down or overexpressed the target gene and phosphorylation inhibition of Smad verified the influence of downstream molecules on the abundance of miR-27b-3p and fibrogenic differentiation of FAPs. RESULT: FAPs derived from a mouse model with sciatic nerves denervated exhibited a progressively worsening fibrotic phenotype over time. Introducing agomiR-27b-3p effectively suppressed fibrosis both in vitro and in vivo. MiR-27b-3p targeted Transforming Growth Factor Beta Receptor 1 (TGF-ßR1) and the abundance of miR-27b-3p was negatively regulated by TGF-ßR1/Smad. CONCLUSION: miR-27b-3p targeting the TGF-ßR1/Smad pathway is a novel mechanism for regulating fibrogenic differentiation of FAPs. Increasing abundance of miR-27b-3p, suppressing expression of TGF-ßR1 and inhibiting phosphorylation of smad3 presented potential strategies for treating fibrosis in chronic skeletal muscle injury.


Fibrosis , MicroRNAs , Muscle, Skeletal , Signal Transduction , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mice , Chronic Disease , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Mice, Inbred C57BL , Smad Proteins/metabolism , Smad Proteins/genetics , Male , Disease Models, Animal , Cell Differentiation , Sciatic Nerve/injuries
2.
FASEB J ; 38(9): e23622, 2024 May 15.
Article En | MEDLINE | ID: mdl-38703029

Endometriosis (EMs)-related infertility commonly has decreased endometrial receptivity and normal decidualization is the basis for establishing and maintaining endometrial receptivity. However, the potential molecular regulatory mechanisms of impaired endometrial decidualization in patients with EMs have not been fully clarified. We confirmed the existence of reduced endometrial receptivity in patients with EMs by scanning electron microscopy and quantitative real-time PCR. Here we identified an lncRNA, named BMPR1B-AS1, which is significantly downregulated in eutopic endometrium in EMs patients and plays an essential role in decidual formation. Furthermore, RNA pull-down, mass spectrometry, RNA immunoprecipitation, and rescue analyses revealed that BMPR1B-AS1 positively regulates decidual formation through interaction with the RNA-binding protein insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2). Downregulation of IGF2BP2 led to a decreased stability of BMPR1B-AS1 and inhibition of activation of the SMAD1/5/9 pathway, an inhibitory effect which diminished decidualization in human endometrial stromal cells (hESCs) decidualization. In conclusion, our identified a novel regulatory mechanism in which the IGF2BP2-BMPR1B-AS1-SMAD1/5/9 axis plays a key role in the regulation of decidualization, providing insights into the potential link between abnormal decidualization and infertility in patients with EMs, which will be of clinical significance for the management and treatment of infertility in patients with EMs.


Endometriosis , RNA, Long Noncoding , RNA-Binding Proteins , Adult , Female , Humans , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Decidua/metabolism , Decidua/pathology , Endometriosis/metabolism , Endometriosis/genetics , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Infertility, Female/metabolism , Infertility, Female/genetics , Infertility, Female/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Stromal Cells/metabolism , Smad Proteins , Young Adult
3.
Pak J Pharm Sci ; 37(1): 33-41, 2024 Jan.
Article En | MEDLINE | ID: mdl-38741398

The objective of this research is to assess how salvianolate impacts inflammation and oxidative stress in a laboratory setting, as well as to investigate the underlying mechanisms. HK-2 cells were subjected to different treatments, including normal glucose, mannitol, high glucose and high glucose plus salvianolate. Cell proliferation, death, MDA levels, IL-1ß, IL-6, TNF-α, MCP-1 concentrations, ROS levels, MMP, MPTP and ATP levels were assessed using various kits. The protein expressions of NOX4, TGF-ß1, P-Smad2, P-Smad3, Smad4 and Smad7 were ascertained through western blot analysis. Our results indicated salvianolate could reduce the release of IL-1ß, IL-6, TNF-α, as well as MCP-1, alleviate the levels of oxidative stress markers NOX4 and MDA, and improve mitochondrial function by increasing MMP and ATP levels while reducing ROS and MPTP opening. Furthermore, salvianolate inhibited the TGF-ß1/Smad2, Smad3 signaling pathway, suppressed Smad4 expression and increased Smad7 expression. Salvianolate seems to mitigate inflammation and oxidative stress through a variety of mechanisms. These discoveries offer valuable understanding into the possible mechanisms by which salvianolate may be employed in the treatment of diabetic nephropathy.


Glucose , Inflammation , Oxidative Stress , Signal Transduction , Oxidative Stress/drug effects , Signal Transduction/drug effects , Glucose/metabolism , Humans , Cell Line , Inflammation/drug therapy , Inflammation/metabolism , Smad Proteins/metabolism , Plant Extracts/pharmacology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Reactive Oxygen Species/metabolism , Anti-Inflammatory Agents/pharmacology , Cell Proliferation/drug effects
4.
Arch Dermatol Res ; 316(6): 225, 2024 May 24.
Article En | MEDLINE | ID: mdl-38787453

Myosin Va (Myo Va) is one of three protein complexes involved in melanosome transport. In this study, we identified BMP-2 as an up-regulator of Myo Va expression using 2-methyl-naphtho[1,2,3-de]quinolin-8-one (MNQO). Our results showed that MNQO reduced the mRNA and protein expression of Myo Va and BMP-2 in melanocytes. Knockdown of BMP-2 by siRNA also affected Myo Va mRNA and protein expression, confirming that MNQO regulates Myo Va through BMP-2. Furthermore, phosphorylation of Smad1/5/8 by BMP2 treatment confirmed that the BMP-2/Smad signaling pathway regulates Myo Va expression in Melan-a melanocytes. Smad-binding elements were found in the Myo Va promoter and phosphorylated Smad1/5/8 bind directly to the Myo Va promoter to activate Myo Va transcription and BMP-2 enhances this binding. These findings provide insight into a new role for BMP-2 in Melan-a melanocytes and a mechanism of regulation of Myo Va expression that may be beneficial in the treatment of albinism or hyperpigmentation disorders.


Bone Morphogenetic Protein 2 , Melanocytes , Myosin Heavy Chains , Myosin Type V , Signal Transduction , Myosin Type V/metabolism , Myosin Type V/genetics , Melanocytes/metabolism , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Humans , Smad Proteins/metabolism , Promoter Regions, Genetic/genetics , Phosphorylation , Mice , Animals , Gene Expression Regulation
5.
Am J Chin Med ; 52(3): 753-773, 2024.
Article En | MEDLINE | ID: mdl-38716621

The formation of fibrotic tissue, characterized by the excessive accumulation of extracellular matrix (ECM) components such as collagen and fibronectin, is a normal and crucial stage of tissue repair in all organs. The over-synthesis, deposition, and remodeling of ECM components lead to organ dysfunction, posing a significant medical burden. Berberine, an isoquinoline alkaloid, is commonly used in the treatment of gastrointestinal diseases. With the deepening of scientific research, it has been gradually discovered that berberine also plays an important role in fibrotic diseases. In this review, we systematically introduce the effective role of berberine in fibrosis-related diseases. Specifically, this paper aims to provide a comprehensive review of the therapeutic role of berberine in treating fibrosis in organs such as the heart, liver, lungs, and kidneys. By summarizing its various pathways and mechanisms of action, including the inhibition of the transforming growth factor-[Formula: see text]/Smad signaling pathway, PI3K/Akt signaling pathway, MAPK signaling pathway, RhoA/ROCK signaling, and mTOR/p70S6K signaling pathway, as well as its activation of the Nrf2-ARE signaling pathway, AMPK signaling pathway, phosphorylated Smad 2/3 and Smad 7, and other signaling pathways, this review offers additional evidence to support the treatment of fibrotic diseases.


Berberine , Fibrosis , Signal Transduction , Berberine/pharmacology , Berberine/therapeutic use , Humans , Signal Transduction/drug effects , Smad Proteins/metabolism , Phytotherapy , Animals , Kidney Diseases/drug therapy , Kidney Diseases/etiology
6.
Cell Biol Toxicol ; 40(1): 39, 2024 May 25.
Article En | MEDLINE | ID: mdl-38789630

Hypertrophic scar (HS) is characterized by excessive collagen deposition and myofibroblasts activation. Endothelial-to-mesenchymal transition (EndoMT) and oxidative stress were pivotal in skin fibrosis process. Exosomes derived from adipose tissue-derived stem cells (ADSC-Exo) have the potential to attenuate EndoMT and inhibit fibrosis. The study revealed reactive oxygen species (ROS) levels were increased during EndoMT occurrence of dermal vasculature of HS. The morphology of endothelial cells exposure to H2O2, serving as an in vitro model of oxidative stress damage, transitioned from a cobblestone-like appearance to a spindle-like shape. Additionally, the levels of endothelial markers decreased in H2O2-treated endothelial cell, while the expression of fibrotic markers increased. Furthermore, H2O2 facilitated the accumulation of ROS, inhibited cell proliferation, retarded its migration and suppressed tube formation in endothelial cell. However, ADSC-Exo counteracted the biological effects induced by H2O2. Subsequently, miRNAs sequencing analysis revealed the significance of mir-486-3p in endothelial cell exposed to H2O2 and ADSC-Exo. Mir-486-3p overexpression enhanced the acceleration of EndoMT, its inhibitors represented the attenuation of EndoMT. Meanwhile, the target regulatory relationship was observed between mir-486-3p and Sirt6, whereby Sirt6 exerted its anti-EndoMT effect through Smad2/3 signaling pathway. Besides, our research had successfully demonstrated the impact of ADSC-Exo and mir-486-3p on animal models. These findings of our study collectively elucidated that ADSC-Exo effectively alleviated H2O2-induced ROS and EndoMT by inhibiting the mir-486-3p/Sirt6/Smad axis.


Adipose Tissue , Exosomes , Human Umbilical Vein Endothelial Cells , Hydrogen Peroxide , MicroRNAs , Oxidative Stress , Signal Transduction , Sirtuins , MicroRNAs/metabolism , MicroRNAs/genetics , Humans , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/toxicity , Oxidative Stress/drug effects , Sirtuins/metabolism , Sirtuins/genetics , Signal Transduction/drug effects , Exosomes/metabolism , Exosomes/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Adipose Tissue/metabolism , Reactive Oxygen Species/metabolism , Smad Proteins/metabolism , Epithelial-Mesenchymal Transition/drug effects , Cell Proliferation/drug effects , Smad2 Protein/metabolism , Animals , Stem Cells/metabolism , Stem Cells/drug effects , Cell Movement/drug effects
7.
Biol Pharm Bull ; 47(5): 1008-1020, 2024.
Article En | MEDLINE | ID: mdl-38797693

The dipeptidyl peptidase-4 (DPP-4) inhibitors, a novel anti-diabetic medication family, are renoprotective in diabetes, but a comparable benefit in chronic non-diabetic kidney diseases is still under investigation. This study aimed to elucidate the molecular mechanisms of linagliptin's (Lina) protective role in a rat model of chronic kidney injury caused by tacrolimus (TAC) independent of blood glucose levels. Thirty-two adult male Sprague Dawley rats were equally randomized into four groups and treated daily for 28 d as follows: The control group; received olive oil (1 mL/kg/d, subcutaneously), group 2; received Lina (5 mg/kg/d, orally), group 3; received TAC (1.5 mg/kg/d, subcutaneously), group 4; received TAC plus Lina concomitantly in doses as the same previous groups. Blood and urine samples were collected to investigate renal function indices and tubular injury markers. Additionally, signaling molecules, epithelial-mesenchymal transition (EMT), and fibrotic-related proteins in kidney tissue were assessed by enzyme-linked immunosorbent assay (ELISA) and Western blot analysis, immunohistochemical and histological examinations. Tacrolimus markedly induced renal injury and fibrosis as indicated by renal dysfunction, histological damage, and deposition of extracellular matrix (ECM) proteins. It also increased transforming growth factor ß1 (TGF-ß1), Smad4, p-extracellular signal-regulated kinase (ERK)1/2/ERK1/2, and p-P38/P38 mitogen-activated protein kinase (MAPK) protein levels. These alterations were markedly attenuated by the Lina administration. Moreover, Lina significantly inhibited EMT, evidenced by inhibiting Vimentin and α-smooth muscle actin (α-SMA) and elevating E-cadherin. Furthermore, Lina diminished hypoxia-related protein levels with a subsequent reduction in Snail and Twist expressions. We concluded that Lina may protect against TAC-induced interstitial fibrosis by modulating TGF-ß1 mediated EMT via Smad-dependent and independent signaling pathways.


Epithelial-Mesenchymal Transition , Fibrosis , Linagliptin , Rats, Sprague-Dawley , Tacrolimus , Transforming Growth Factor beta1 , Animals , Linagliptin/pharmacology , Linagliptin/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Male , Tacrolimus/pharmacology , Transforming Growth Factor beta1/metabolism , Signal Transduction/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Rats , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Immunosuppressive Agents/pharmacology
8.
Theriogenology ; 225: 67-80, 2024 Sep 01.
Article En | MEDLINE | ID: mdl-38795512

Here, we examined the effects of the BMP signaling pathway inhibitor LDN-193189 on the pluripotency of porcine embryonic stem cells (ESCs) in the absence of feeder cells using molecular and transcriptomic techniques. Additionally, the effects of some extracellular matrix components on porcine ESC pluripotency were evaluated to develop an optimized and sustainable feeder-free culture system for porcine ESCs. Feeder cells were found to play an important role in supporting the pluripotency of porcine ESCs by blocking trophoblast and mesodermal differentiation through the inhibition of the BMP pathway. Additionally, treatment with LDN-193189, an inhibitor of the BMP pathway, maintained the pluripotency and homogeneity of porcine ESCs for an extended period in the absence of feeder cells by stimulating the secretion of chemokines and suppressing differentiation, based on transcriptome analysis. Conclusively, these results suggest that LDN-193189 could be a suitable replacement for feeder cells in the maintenance of porcine ESC pluripotency during culture. Additionally, these findings contribute to the understanding of pluripotency gene networks and comparative embryogenesis.


Embryonic Stem Cells , Pyrazoles , Signal Transduction , Animals , Swine , Embryonic Stem Cells/drug effects , Signal Transduction/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Bone Morphogenetic Proteins/metabolism , Pluripotent Stem Cells/drug effects , Cell Differentiation/drug effects , Smad Proteins/metabolism , Smad Proteins/genetics , Feeder Cells , Cell Culture Techniques
9.
PLoS Comput Biol ; 20(5): e1012072, 2024 May.
Article En | MEDLINE | ID: mdl-38753874

Cells use signaling pathways to sense and respond to their environments. The transforming growth factor-ß (TGF-ß) pathway produces context-specific responses. Here, we combined modeling and experimental analysis to study the dependence of the output of the TGF-ß pathway on the abundance of signaling molecules in the pathway. We showed that the TGF-ß pathway processes the variation of TGF-ß receptor abundance using Liebig's law of the minimum, meaning that the output-modifying factor is the signaling protein that is most limited, to determine signaling responses across cell types and in single cells. We found that the abundance of either the type I (TGFBR1) or type II (TGFBR2) TGF-ß receptor determined the responses of cancer cell lines, such that the receptor with relatively low abundance dictates the response. Furthermore, nuclear SMAD2 signaling correlated with the abundance of TGF-ß receptor in single cells depending on the relative expression levels of TGFBR1 and TGFBR2. A similar control principle could govern the heterogeneity of signaling responses in other signaling pathways.


Signal Transduction , Transforming Growth Factor beta , Transforming Growth Factor beta/metabolism , Humans , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Smad2 Protein/metabolism , Computational Biology , Models, Biological , Cell Line, Tumor , Smad Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 652-659, 2024 Apr 20.
Article Zh | MEDLINE | ID: mdl-38708497

OBJECTIVE: To investigate the protective effect of arbutin against CCl4-induced hepatic fibrosis in mice and explore the underlying mechanisms. METHODS: Twenty-four C57BL/6 mice were randomly divided into control group, model group, and low- and high-dose arbutin treatment (25 and 50 mg/kg, respectively) groups. Mouse models of liver fibrosis were established by intraperitoneal injection of CCl4, and arbutin was administered daily via gavage for 6 weeks. After the treatments, serum biochemical parameters of the mice were tested, and liver tissues were taken for HE staining, Sirius Red staining and immunohistochemical staining. RT-qPCR was used to detect the mRNA levels of α-SMA, Pdgfb, Col1α1, Timp-1, Ccl2 and Tnf-a, and Western blotting was performed to detect α-SMA protein expression in the liver tissues. In the cell experiment, the effect of arbutin treatment for 24 h on THP-1 and RAW264.7 cell migration and recruitment was examined using Transwell migration assay and DAPI staining; The changes in protein levels of Akt, p65, Smad3, p-Akt, p-p65, p-Smad3 and α-SMA in arbutintreated LX-2 cells were detected with Western blotting. RESULTS: Arbutin treatment significantly lowered serum alanine aminotransferase and aspartate aminotransferase levels, alleviated liver tissue damage and collagen deposition, and reduced macrophage infiltration and α-SMA protein expression in the liver of the mouse models (P < 0.05 or 0.001). Arbutin treatment also significantly reduced CCl4-induced elevation of a-SMA, Pdgfb, Col1α1, Timp-1, Ccl2 and Tnf-a mRNA levels in mice (P < 0.05). In the cell experiment, arbutin treatment obviously inhibited migration and recruitment of THP-1 and RAW264.7 cells and lowered the phosphorylation levels of Akt, p65 and Smad3 and the protein expression level of α-SMA in LX-2 cells. CONCLUSION: Arbutin ameliorates liver inflammation and fibrosis in mice by inhibiting hepatic stellate cell activation via reducing macrophage recruitment and infiltration and suppressing activation of the Akt/NF-κB and Smad signaling pathways.


Arbutin , Liver Cirrhosis , Macrophages , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Male , Mice , Arbutin/pharmacology , Arbutin/therapeutic use , Carbon Tetrachloride , Cell Movement/drug effects , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Liver/metabolism , Liver/pathology , Liver/drug effects , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Macrophages/metabolism , Macrophages/drug effects , Mice, Inbred C57BL , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Smad Proteins/metabolism
11.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 95-101, 2024 Mar 31.
Article En | MEDLINE | ID: mdl-38650149

Osteoporosis is a common chronic bone disorder in postmenopausal women. Ginsenosides are primary active components in ginseng and the effects of various ginsenoside variants in osteoporosis treatment have been widely revealed. We planned to explore the impact of ginsenoside Rc on bone resorption in an osteoporosis rat model. We used ovariectomized rats to assess the potential impact of ginsenoside Rc on osteoporosis. µ-CT was implemented for analyzing the microstructure of the distal left femur in rats. H&E staining together with Masson staining were applied for bone histomorphometry evaluation. ELISA kits were implemented to detect serum concentrations of TRACP-5b, OCN, CTX, as well as PINP. Ginsenoside Rc treatment lessened the serum levels of TRACP-5b as well as CTX, while increasing serum levels of OCN, and PINP of OVX rats. Moreover, we found that ginsenoside Rc contributed to the synthesis of type I collagen via increasing Col1a1 and Col1a2 levels in femur tissues of ovariectomized rats. Our findings also revealed that ginsenoside Rc activated the TGF-ß/Smad pathway by increasing TGF-ß as well as phosphorylated Smad2/3 protein levels. Ginsenoside Rc alleviates osteoporosis in rats through promoting the TGF-ß/Smad pathway.


Ginsenosides , Osteoporosis , Ovariectomy , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Animals , Female , Osteoporosis/drug therapy , Osteoporosis/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Femur/drug effects , Femur/metabolism , Femur/pathology , Smad Proteins/metabolism , Rats , Collagen Type I/metabolism , X-Ray Microtomography , Tartrate-Resistant Acid Phosphatase/metabolism , Osteocalcin/metabolism , Osteocalcin/blood , Disease Models, Animal , Procollagen/metabolism , Procollagen/blood
12.
Int Immunopharmacol ; 133: 112131, 2024 May 30.
Article En | MEDLINE | ID: mdl-38669945

BACKGROUND: Osthole is a natural active ingredient extracted from the traditional Chinese medicine Cnidium monnieri. It has been demonstrated to have anti-inflammatory, anti-fibrotic, and anti-hyperglycemic properties. However, its effect on diabetic kidney disease (DKD) remains uncertain. This study aims to assess the preventive and therapeutic effects of osthole on DKD and investigate its underlying mechanisms. METHODS: A streptozotocin/high-fat and high-sucrose diet induced Type 2 diabetic rat model was established. Metformin served as the positive drug control. Diabetic rats were treated with metformin or three different doses of osthole for 8 weeks. Throughout the treatment period, the progression of DKD was assessed by monitoring increases in urinary protein, serum creatinine, urea nitrogen, and uric acid, along with scrutinizing kidney pathology. Enzyme-linked immunosorbent assay (ELISA) was employed to detect inflammatory factors and oxidative stress levels. At the same time, immunohistochemical staining was utilized to evaluate changes in alpha-smooth muscle actin, fibronectin, E-cadherin, and apoptosis. The alterations in TGF-ß1/Smads signaling pathway were ascertained through western blot and immunofluorescence. Furthermore, we constructed a high glucose-stimulated HBZY-1 cells model to uncover its molecular protective mechanism. RESULTS: Osthole significantly reduced fasting blood glucose, insulin resistance, serum creatinine, uric acid, blood urea nitrogen, urinary protein excretion, and glomerular mesangial matrix deposition in diabetic rats. Additionally, significant improvements were observed in inflammation, oxidative stress, apoptosis, and fibrosis levels. The increase of ROS, apoptosis and hypertrophy in HBZY-1 cells induced by high glucose was reduced by osthole. Immunofluorescence and western blot results demonstrated that osthole down-regulated the TGF-ß1/Smads signaling pathway and related protein expression. CONCLUSION: Our findings indicate that osthole exhibits potential preventive and therapeutic effects on DKD. It deserves further investigation as a promising drug for preventing and treating DKD.


Coumarins , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Oxidative Stress , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta1 , Animals , Coumarins/pharmacology , Coumarins/therapeutic use , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Diabetes Mellitus, Experimental/drug therapy , Male , Transforming Growth Factor beta1/metabolism , Rats , Smad Proteins/metabolism , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Cell Line , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Humans , Inflammation/drug therapy
13.
Oral Oncol ; 152: 106798, 2024 May.
Article En | MEDLINE | ID: mdl-38615583

Acquired radio-resistance is thought to be one of the main causes of recurrent metastasis after failure of nasopharyngeal carcinoma (NPC) radiotherapy, which may be related to X-ray-induced epithelial-mesenchymal transition (EMT) activation. The circadian clock gene, BMAL1, has been shown to correlate with the sensitivity of NPCs to radiotherapy, but the specific mechanism has not been reported. NPC cells were irradiated by conventional fractionation to generate radiotherapy-resistant cells. NPC cells with BMAL1 gene stabilization/overexpression and interference were obtained by lentiviral transfection. Western blotting, colony formation analysis, cell counting kit-8 assays, wound-healing tests, Transwell assays, flow cytometry, the EDU method, nuclear plasma separation experiments, HE staining, immunohistochemical staining and TUNEL staining were performed to explore the influence and molecular mechanism of the circadian clock gene, BMAL1, on NPC-acquired radio-resistance and EMT through in vitro and in vivo experiments. The results indicated that there was a gradual downregulation of BMAL1 gene protein expression during the routine dose induction of radio-resistance in NPC cells. EMT activation was present in the radiation-resistant cell line 5-8FR, and was accompanied by the significant enhancement of proliferation, migration and invasion. The BMAL1 gene significantly increased the radiosensitivity of the radiation-resistant cell line 5-8FR and reversed the acquired radio-resistance of NPCs, which was accomplished by inhibiting the TGF-ß1/Smads/Snail1 axis-mediated EMT.


ARNTL Transcription Factors , Epithelial-Mesenchymal Transition , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Radiation Tolerance , Snail Family Transcription Factors , Transforming Growth Factor beta1 , Humans , Snail Family Transcription Factors/metabolism , ARNTL Transcription Factors/metabolism , ARNTL Transcription Factors/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Transforming Growth Factor beta1/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/radiotherapy , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/genetics , Cell Line, Tumor , Animals , Mice , Smad Proteins/metabolism , Mice, Nude , Circadian Clocks , Male
14.
Mol Med ; 30(1): 52, 2024 Apr 19.
Article En | MEDLINE | ID: mdl-38641575

BACKGROUND: Skin fibrosis affects the normal function of the skin. TGF-ß1 is a key cytokine that affects organ fibrosis. The latency-associated peptide (LAP) is essential for TGF-ß1 activation. We previously constructed and prepared truncated LAP (tLAP), and confirmed that tLAP inhibited liver fibrosis by affecting TGF-ß1. SPACE peptide has both transdermal and transmembrane functions. SPACE promotes the delivery of macromolecules through the stratum corneum into the dermis. This study aimed to alleviate skin fibrosis through the delivery of tLAP by SPACE. METHODS: The SPACE-tLAP (SE-tLAP) recombinant plasmid was constructed. SE-tLAP was purified by nickel affinity chromatography. The effects of SE-tLAP on the proliferation, migration, and expression of fibrosis-related and inflammatory factors were evaluated in TGF-ß1-induced NIH-3T3 cells. F127-SE-tLAP hydrogel was constructed by using F127 as a carrier to load SE-tLAP polypeptide. The degradation, drug release, and biocompatibility of F127-SE-tLAP were evaluated. Bleomycin was used to induce skin fibrosis in mice. HE, Masson, and immunohistochemistry were used to observe the skin histological characteristics. RESULTS: SE-tLAP inhibited the proliferation, migration, and expression of fibrosis-related and inflammatory factors in NIH-3T3 cells. F127-SE-tLAP significantly reduced ECM production, collagen deposition, and fibrotic pathological changes, thereby alleviating skin fibrosis. CONCLUSION: F127-SE-tLAP could increase the transdermal delivery of LAP, reduce the production and deposition of ECM, inhibit the formation of dermal collagen fibers, and alleviate the progression of skin fibrosis. It may provide a new idea for the therapy of skin fibrosis.


Polyethylenes , Polypropylenes , Skin Diseases , Transforming Growth Factor beta , Animals , Mice , Bleomycin/adverse effects , Collagen/metabolism , Fibrosis/drug therapy , Hydrogels/chemistry , Hydrogels/pharmacology , Polyethylenes/pharmacology , Polypropylenes/pharmacology , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Skin Diseases/chemically induced , Skin Diseases/drug therapy , Skin Diseases/metabolism , Smad Proteins/drug effects , Smad Proteins/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology
15.
Pharmacol Res ; 204: 107189, 2024 Jun.
Article En | MEDLINE | ID: mdl-38649124

Spinal cord injury (SCI) is a complex problem in modern medicine. Fibroblast activation and fibroscarring after SCI impede nerve recovery. Non-coding RNA plays an important role in the progression of many diseases, but the study of its role in the progression of spinal fibrosis is still emerging. Here, we investigated the function of circular RNAs, specifically antisense to the cerebellar degeneration-related protein 1 (CDR1as), in spinal fibrosis and characterized its molecular mechanism and pathophysiology. The presence of CDR1as in the spinal cord was verified by sequencing and RNA expression assays. The effects of inhibition of CDR1as on scar formation, inflammation and nerve regeneration after spinal cord injury were investigated in vivo and in vitro. Further, gene expression of miR-7a-5p and protein expression of transforming Growth Factor Beta Receptor II (TGF-ßR2) were measured to evaluate their predicted interactions with CDR1as. The regulatory effects and activation pathways were subsequently verified by miR-7a-5p inhibitor and siCDR1as. These results indicate that CDR1as/miR-7a-5p/TGF-ßR2 interactions may exert scars and nerves functions and suggest potential therapeutic targets for treating spinal fibrotic diseases.


Fibrosis , MicroRNAs , RNA, Circular , RNA, Long Noncoding , Signal Transduction , Spinal Cord Injuries , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Smad Proteins/metabolism , Smad Proteins/genetics , Nerve Regeneration , Female , Male , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Mice, Inbred C57BL , Mice , Recovery of Function
16.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 158-163, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38678608

Dermal papilla cell (DPC) belongs to a specialized mesenchymal stem cell for hair follicle regeneration. Maintaining the ability of DPCs to stimulate hair in vitro culture is important for hair follicle morphogenesis and regeneration. As the third generation of platelet concentrate, injectable platelet-rich fibrin (i-PRF) is a novel biomaterial containing many growth factors and showing promising effects on tissue reconstruction. We aimed to explore the influences of i-PRF on the proliferative, migratory, as well as trichogenic ability of DPCs and compared the effects of i-PRF and platelet-rich plasma (PRP), the first generation of platelet concentrate. Both PRP and i-PRF facilitated DPCs proliferation, and migration, along with trichogenic inductivity as well as stimulated the TGF-ß/Smad pathway, while the impacts of i-PRF were more significant than PRP. A small molecule inhibitor of TGF-beta receptor I, Galunisertib, was also applied to treat DPCs, and it rescued the impacts of i-PRF on the proliferative, migratory, trichogenic inductivity, and proteins-associated with TGF-ß/Smad pathway in DPCs. These findings revealed that i-PRF had better effects than PRP in enhancing the proliferative, migratory, and hair-inducing abilities of DPCs by the TGF-ß/Smad pathway, which indicated the beneficial role of i-PRF in hair follicle regeneration.


Cell Movement , Cell Proliferation , Hair Follicle , Platelet-Rich Fibrin , Signal Transduction , Smad Proteins , Transforming Growth Factor beta , Signal Transduction/drug effects , Cell Proliferation/drug effects , Transforming Growth Factor beta/metabolism , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/cytology , Smad Proteins/metabolism , Humans , Platelet-Rich Fibrin/metabolism , Cell Movement/drug effects , Dermis/cytology , Dermis/metabolism , Dermis/drug effects , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Platelet-Rich Plasma/metabolism , Injections
17.
Reprod Biol ; 24(2): 100882, 2024 Jun.
Article En | MEDLINE | ID: mdl-38604016

This study aims to elucidate the effects of Platelet-rich plasma (PRP) in fibrosis development in intrauterine adhesion (IUA), and the associated underlying mechanisms are also explored, which are expected to be a potential therapeutic scheme for IUA. In this research, PRP was obtained and prepared from the peripheral venous blood of rats. A rat model was induced by mechanical injury. Further, PRP was directly injected into the uterus for treatment. The appearance and shape of the uterus were assessed based on the tissues harvested. The fibrosis biomarker levels were analyzed. The transforming growth factor beta 1 (TGF-ß1) and Mothers against decapentaplegic homolog 7 (Smad7) levels, the phosphorylation of Smad2 (p-Smad2), and the phosphorylation of Smad3 (p-Smad3) were analyzed, and the molecular mechanism was investigated by rescue experiments. It was found that PRP improved the appearance and shape of the uterus in IUA and increased endometrial thickness and gland numbers. The administration of PRP resulted in a decrease in the expressions of fibrosis markers including collagen I, α-SMA, and fibronectin. Furthermore, PRP increased Smad7 levels and decreased TGF-ß1 levels, p-Smad2, and p-Smad3. Meanwhile, administration of TGF-ß1 activator reversed the therapeutic effects of PRP in IUA. Collectively, the intrauterine infusion of PRP can promote endometrial damage recovery and improve endometrial fibrosis via the TGF-ß1/Smad pathway. Hence, PRP can be a potential therapeutic strategy for IUA.


Fibrosis , Platelet-Rich Plasma , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta1 , Uterine Diseases , Uterus , Animals , Female , Transforming Growth Factor beta1/metabolism , Rats , Tissue Adhesions/metabolism , Uterine Diseases/therapy , Uterine Diseases/metabolism , Signal Transduction/drug effects , Uterus/metabolism , Disease Models, Animal , Smad Proteins/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism
18.
Mol Biol Rep ; 51(1): 529, 2024 Apr 18.
Article En | MEDLINE | ID: mdl-38637422

BACKGROUND: TGF-ß1 and SMAD3 are particularly pathogenic in the progression of renal fibrosis. AIM: This study aimed to evaluate the kidney protective potentials of silymarin (SM) and exosomes of mesenchymal stem cells against the nephrotoxin thioacetamide (TAA) in rats. METHODS: 32 female rats were randomly assigned into four groups: the control group, the TAA group, the TAA + SM group, and the TAA + Exosomes group. The kidney homogenates from all groups were examined for expression levels of TGF-ß receptors I and II using real-time PCR, expression levels of collagen type I and CTGF proteins using ELISA, and the expression levels of nuclear SMAD2/3/4, cytoplasmic SMAD2/3, and cytoplasmic SMAD4 proteins using the western blot technique. RESULTS: Compared to the control group, the injection of TAA resulted in a significant increase in serum levels of urea and creatinine, gene expression levels of TßRI and TßRII, protein expression levels of both collagen I and CTGF proteins, cytoplasmic SMAD2/3 complex, and nuclear SMAD2/3/4 (p-value < 0.0001), with significantly decreased levels of the co-SMAD partner, SMAD4 (p-value < 0.0001). Those effects were reversed considerably in both treatment groups, with the superiority of the exosomal treatment regarding the SMAD proteins and the expression levels of the TßRI gene, collagen I, and CTGF proteins returning to near-control values (p-value > 0.05). CONCLUSION: Using in vitro and in vivo experimental approaches, the research discovered a reno-protective role of silymarin and exosomes of BM-MSCs after thioacetamide-induced renal fibrosis in rats, with the advantage of exosomes.


Exosomes , Kidney Diseases , Silymarin , Rats , Female , Animals , Transforming Growth Factor beta/metabolism , Thioacetamide/toxicity , Thioacetamide/metabolism , Silymarin/pharmacology , Exosomes/metabolism , Fibrosis , Transforming Growth Factor beta1/metabolism , Kidney Diseases/pathology , Collagen Type I/metabolism , Smad Proteins/metabolism
19.
Life Sci ; 346: 122626, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38614295

AIM: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive condition with unknown aetiology that causes the lung parenchyma to scar incessantly, lowering the quality of life and hastening death. In this investigation, we studied the anti-fibrotic activity of Geneticin (a derivative of gentamycin) using in vitro and in vivo models. MAIN METHODS: The TGF-ß-mediated differentiation model was adopted to investigate (fibrotic marker's levels/expression) the anti-fibrotic activity of geneticin (GNC) in in-vitro scenarios (LL29 and DHLF cells). In vivo, the bleomycin (BLM)-induced pulmonary fibrosis model was employed by administering BLM intratracheally. Post 14 days of BLM administration, animals were treated with geneticin (6.25, 12.5, and 25 mg·kg-1) for another 14 days, and their therapeutic effect was investigated using a spectrum of techniques. KEY FINDINGS: RTqPCR and western-blot results revealed that geneticin treatment significantly attenuated the TGF-ß/BLM mediated fibrotic cascade of markers in both in-vitro and in-vivo models respectively. Further, the BLM-induced pulmonary fibrosis model revealed, that geneticin dose-dependently reduced the BLM-induced inflammatory cell infiltrations, and thickness of the alveoli walls, improved the structural distortion of the lung, and aided in improving the survival rate of the rats. Picrosirus and Masson's trichrome staining indicated that geneticin therapy reduced collagen deposition and, as a result, lung functional characteristics were improved as assessed by flexivent. Mechanistic studies have shown that geneticin reduced fibrosis by attenuating the TGF-ß/Smad through modulating the AMPK/SIRT1 signaling. SIGNIFICANCE: These findings suggest that geneticin may be a promising therapeutic agent for the treatment of pulmonary fibrosis in clinical settings.


AMP-Activated Protein Kinases , Bleomycin , Pulmonary Fibrosis , Signal Transduction , Sirtuin 1 , Transforming Growth Factor beta , Animals , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/chemically induced , Rats , Sirtuin 1/metabolism , Sirtuin 1/genetics , Male , Bleomycin/toxicity , AMP-Activated Protein Kinases/metabolism , Smad Proteins/metabolism , Rats, Sprague-Dawley , Disease Models, Animal
20.
Int J Biol Macromol ; 264(Pt 1): 130502, 2024 Apr.
Article En | MEDLINE | ID: mdl-38428779

Hepatic stellate cell (HSC) activation is a crucial step in the development of liver fibrosis. Previous studies have shown that antler stem cells (AnSCs) inhibited HSC activation, suggesting that this may be achieved through secreting or releasing peptides. This study aimed to investigate whether AnSC-derived peptides (AnSC-P) could reduce liver fibrosis. The results showed that AnSC-P effectively reduced liver fibrosis in rats. Furthermore, we found that thymosin ß10 (Tß-10) was rich in AnSC-P, which may be the main component of AnSC-P contributing to the reduction in liver fibrosis. A further study showed that Tß-10 reduced liver fibrosis in rats, with a reduction in HYP and MDA levels in the liver tissues, a decrease in the serum levels of ALP, ALT, AST, and TBIL and an increase in TP and ALB. Moreover, Tß-10 decreased the expression levels of the genes related to the TGF-ß/SMAD signaling pathway in vivo. In addition, Tß-10 also inhibited TGF-ß1-induced HSC activation and decreased the expression levels of the TGF-ß/SMAD signaling pathway-related genes in HSCs in vitro. In conclusion, antler Tß-10 is a potential drug candidate for the treatment of liver fibrosis, the effect of which may be achieved via inhibition of the TGFß/SMAD signaling pathway.


Antlers , Thymosin , Transforming Growth Factor beta1 , Rats , Animals , Transforming Growth Factor beta1/metabolism , Antlers/metabolism , Smad Proteins/metabolism , Hepatic Stellate Cells , Liver Cirrhosis/chemically induced , Transforming Growth Factor beta/metabolism
...