Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 986
Filter
1.
FASEB J ; 38(13): e23749, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38953707

ABSTRACT

Pulmonary fibrosis is a formidable challenge in chronic and age-related lung diseases. Myofibroblasts secrete large amounts of extracellular matrix and induce pro-repair responses during normal wound healing. Successful tissue repair results in termination of myofibroblast activity via apoptosis; however, some myofibroblasts exhibit a senescent phenotype and escape apoptosis, causing over-repair that is characterized by pathological fibrotic scarring. Therefore, the removal of senescent myofibroblasts using senolytics is an important method for the treatment of pulmonary fibrosis. Procyanidin C1 (PCC1) has recently been discovered as a senolytic compound with very low toxicity and few side effects. This study aimed to determine whether PCC1 could improve lung fibrosis by promoting apoptosis in senescent myofibroblasts and to investigate the mechanisms involved. The results showed that PCC1 attenuates bleomycin (BLM)-induced pulmonary fibrosis in mice. In addition, we found that PCC1 inhibited extracellular matrix deposition and promoted the apoptosis of senescent myofibroblasts by increasing PUMA expression and activating the BAX signaling pathway. Our findings represent a new method of pulmonary fibrosis management and emphasize the potential of PCC1 as a senotherapeutic agent for the treatment of pulmonary fibrosis, providing hope for patients with pulmonary fibrosis worldwide. Our results advance our understanding of age-related diseases and highlight the importance of addressing cellular senescence in treatment.


Subject(s)
Bleomycin , Catechin , Cellular Senescence , Mice, Inbred C57BL , Myofibroblasts , Pulmonary Fibrosis , Animals , Bleomycin/toxicity , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Mice , Cellular Senescence/drug effects , Catechin/pharmacology , Catechin/analogs & derivatives , Proanthocyanidins/pharmacology , Apoptosis/drug effects , Male , Biflavonoids/pharmacology , Signal Transduction/drug effects
2.
Respir Res ; 25(1): 242, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38877465

ABSTRACT

BACKGROUND: Silicosis represents a paramount occupational health hazard globally, with its incidence, morbidity, and mortality on an upward trajectory, posing substantial clinical dilemmas due to limited effective treatment options available. Trigonelline (Trig), a plant alkaloid extracted mainly from coffee and fenugreek, have diverse biological properties such as protecting dermal fibroblasts against ultraviolet radiation and has the potential to inhibit collagen synthesis. However, it's unclear whether Trig inhibits fibroblast activation to attenuate silicosis-induced pulmonary fibrosis is unclear. METHODS: To evaluate the therapeutic efficacy of Trig in the context of silicosis-related pulmonary fibrosis, a mouse model of silicosis was utilized. The investigation seeks to elucidated Trig's impact on the progression of silica-induced pulmonary fibrosis by evaluating protein expression, mRNA levels and employing Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius Red staining. Subsequently, we explored the mechanism underlying of its functions. RESULTS: In vivo experiment, Trig has been demonstrated the significant efficacy in mitigating SiO2-induced silicosis and BLM-induced pulmonary fibrosis, as evidenced by improved histochemical staining and reduced fibrotic marker expressions. Additionally, we showed that the differentiation of fibroblast to myofibroblast was imped in Trig + SiO2 group. In terms of mechanism, we obtained in vitro evidence that Trig inhibited fibroblast-to-myofibroblast differentiation by repressing TGF-ß/Smad signaling according to the in vitro evidence. Notably, our finding indicated that Trig seemed to be safe in mice and fibroblasts. CONCLUSION: In summary, Trig attenuated the severity of silicosis-related pulmonary fibrosis by alleviating the differentiation of myofibroblasts, indicating the development of novel therapeutic approaches for silicosis fibrosis.


Subject(s)
Alkaloids , Cell Differentiation , Fibroblasts , Mice, Inbred C57BL , Myofibroblasts , Pulmonary Fibrosis , Silicon Dioxide , Silicosis , Animals , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/prevention & control , Alkaloids/pharmacology , Silicon Dioxide/toxicity , Mice , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Cell Differentiation/drug effects , Silicosis/pathology , Silicosis/metabolism , Silicosis/drug therapy , Male
3.
Stem Cell Res Ther ; 15(1): 166, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38867276

ABSTRACT

BACKGROUND: Hypertrophic scarring results from myofibroblast differentiation and persistence during wound healing. Currently no effective treatment for hypertrophic scarring exists however, autologous fat grafting has been shown to improve scar elasticity, appearance, and function. The aim of this study was to understand how paracrine factors from adipose tissues and adipose-derived stromal cells (ADSC) affect fibroblast to myofibroblast differentiation. METHODS: The transforming growth factor-ß1 (TGF-ß1) induced model of myofibroblast differentiation was used to test the effect of conditioned media from adipose tissue, ADSC or lipid on the proportion of fibroblasts and myofibroblasts. RESULTS: Adipose tissue conditioned media inhibited the differentiation of fibroblasts to myofibroblasts but this inhibition was not observed following treatment with ADSC or lipid conditioned media. Hepatocyte growth factor (HGF) was readily detected in the conditioned medium from adipose tissue but not ADSC. Cells treated with HGF, or fortinib to block HGF, demonstrated that HGF was not responsible for the inhibition of myofibroblast differentiation. Conditioned media from adipose tissue was shown to reduce the proportion of myofibroblasts when added to fibroblasts previously treated with TGF-ß1, however, conditioned media treatment was unable to significantly reduce the proportion of myofibroblasts in cell populations isolated from scar tissue. CONCLUSIONS: Cultured ADSC or adipocytes have been the focus of most studies, however, this work highlights the importance of considering whole adipose tissue to further our understanding of fat grafting. This study supports the use of autologous fat grafts for scar treatment and highlights the need for further investigation to determine the mechanism.


Subject(s)
Adipose Tissue , Cell Differentiation , Hepatocyte Growth Factor , Myofibroblasts , Transforming Growth Factor beta1 , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Myofibroblasts/cytology , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Humans , Hepatocyte Growth Factor/pharmacology , Hepatocyte Growth Factor/metabolism , Paracrine Communication/drug effects , Phenotype , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/cytology , Adipocytes/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Stromal Cells/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects
4.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(3): 505-511, 2024 Jun 18.
Article in Chinese | MEDLINE | ID: mdl-38864137

ABSTRACT

OBJECTIVE: To investigate the effect of tofacitinib, a pan-Janus kinase (JAK) inhibitor, on transforming growth factor-beta 1 (TGF-ß1)-induced fibroblast to myofibroblast transition (FMT) and to explore its mechanism. To provide a theoretical basis for the clinical treatment of connective tissue disease-related interstitial lung disease (CTD-ILD). METHODS: (1) Human fetal lung fibroblast 1 (HFL-1) were cultured in vitro, and 6 groups were established: DMSO blank control group, TGF-ß1 induction group, and TGF-ß1 with different concentrations of tofacitinib (0.5, 1.0, 2.0, 5.0 µmol/L) drug intervention experimental groups. CCK-8 was used to measure the cell viability, and wound-healing assay was performed to measure cell migration ability. After 48 h of combined treatment, quantitative real-time PCR (RT-PCR) and Western blotting were used to detect the gene and protein expression levels of α-smooth muscle actin (α-SMA), fibronectin (FN), and collagen type Ⅰ (COL1). (2) RT-PCR and enzyme-linked immunosorbnent assay (ELISA) were used to detect the interleukin-6 (IL-6) gene and protein expression changes, respectively. (3) DMSO carrier controls, 1.0 µmol/L and 5.0 µmol/L tofacitinib were added to the cell culture media of different groups for pre-incubation for 30 min, and then TGF-ß1 was added to treat for 1 h, 6 h and 24 h. The phosphorylation levels of Smad2/3 and signal transducer and activator of transcription 3 (STAT3) protein were detected by Western blotting. RESULTS: (1) Tofacitinib inhibited the viability and migration ability of HFL-1 cells after TGF-ß1 induction. (2) The expression of α-SMA, COL1A1 and FN1 genes of HFL-1 in the TGF-ß1-induced groups was significantly up-regulated compared with the blank control group (P < 0.05). Compared with the TGF-ß1 induction group, α-SMA expression in the 5.0 µmol/L tofacitinib intervention group was significantly inhi-bited (P < 0.05). Compared with the TGF-ß1-induced group, FN1 gene was significantly inhibited in each intervention group at a concentration of 0.5-5.0 µmol/L (P < 0.05). Compared with the TGF-ß1-induced group, the COL1A1 gene expression in each intervention group did not change significantly. (3) Western blotting results showed that the protein levels of α-SMA and FN1 in the TGF-ß1-induced group were significantly higher than those in the control group (P < 0.05), and there was no significant difference in the expression of COL1A1. Compared with the TGF-ß1-induced group, the α-SMA protein level in the intervention groups with different concentrations decreased. And the differences between the TGF-ß1-induced group and 2.0 µmol/L or 5.0 µmol/L intervention groups were statistically significant (P < 0.05). Compared with the TGF-ß1-induced group, the FN1 protein levels in the intervention groups with different concentrations showed a downward trend, but the difference was not statistically significant. There was no difference in COL1A1 protein expression between the intervention groups compared with the TGF-ß1-induced group. (4) After TGF-ß1 acted on HFL-1 cells for 48 h, the gene expression of the IL-6 was up-regulated and IL-6 in culture supernatant was increased, the intervention with tofacitinib partly inhibited the TGF-ß1-induced IL-6 gene expression and IL-6 in culture supernatant. TGF-ß1 induced the increase of Smad2/3 protein phosphorylation in HFL-1 cells for 1 h and 6 h, STAT3 protein phosphorylation increased at 1 h, 6 h and 24 h, the pre-intervention with tofacitinib inhibited the TGF-ß1-induced Smad2/3 phosphorylation at 6 h and inhibited TGF-ß1-induced STAT3 phosphorylation at 1 h, 6 h and 24 h. CONCLUSION: Tofacitinib can inhibit the transformation of HFL-1 cells into myofibroblasts induced by TGF-ß1, and the mechanism may be through inhibiting the classic Smad2/3 pathway as well as the phosphorylation of STAT3 induced by TGF-ß1, thereby protecting the disease progression of pulmonary fibrosis.


Subject(s)
Fibroblasts , Lung , Myofibroblasts , Piperidines , Pyrimidines , STAT3 Transcription Factor , Signal Transduction , Transforming Growth Factor beta1 , Humans , Pyrimidines/pharmacology , Piperidines/pharmacology , STAT3 Transcription Factor/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Transforming Growth Factor beta1/metabolism , Myofibroblasts/metabolism , Myofibroblasts/cytology , Myofibroblasts/drug effects , Lung/cytology , Signal Transduction/drug effects , Fibronectins/metabolism , Cell Movement/drug effects , Pyrroles/pharmacology , Actins/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Janus Kinases/metabolism , Cell Survival/drug effects , Smad2 Protein/metabolism , Lung Diseases, Interstitial/metabolism , Interleukin-6/metabolism , Smad3 Protein/metabolism , Cells, Cultured
5.
Cells ; 13(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38920695

ABSTRACT

Aberrant sialylation with overexpression of the homopolymeric glycan polysialic acid (polySia) was recently reported in fibroblasts from fibrotic skin lesions. Yet, whether such a rise in polySia levels or sialylation in general may be functionally implicated in profibrotic activation of fibroblasts and their transition to myofibroblasts remains unknown. Therefore, we herein explored whether inhibition of sialylation could interfere with the process of skin fibroblast-to-myofibroblast transition induced by the master profibrotic mediator transforming growth factor ß1 (TGFß1). Adult human skin fibroblasts were pretreated with the competitive pan-sialyltransferase inhibitor 3-Fax-peracetyl-Neu5Ac (3-Fax) before stimulation with recombinant human TGFß1, and then analyzed for polySia expression, cell viability, proliferation, migratory ability, and acquisition of myofibroblast-like morphofunctional features. Skin fibroblast stimulation with TGFß1 resulted in overexpression of polySia, which was effectively blunted by 3-Fax pre-administration. Pretreatment with 3-Fax efficiently lessened TGFß1-induced skin fibroblast proliferation, migration, changes in cell morphology, and phenotypic and functional differentiation into myofibroblasts, as testified by a significant reduction in FAP, ACTA2, COL1A1, COL1A2, and FN1 gene expression, and α-smooth muscle actin, N-cadherin, COL1A1, and FN-EDA protein levels, as well as a reduced contractile capability. Moreover, skin fibroblasts pre-administered with 3-Fax displayed a significant decrease in Smad3-dependent canonical TGFß1 signaling. Collectively, our in vitro findings demonstrate for the first time that aberrant sialylation with increased polySia levels has a functional role in skin fibroblast-to-myofibroblast transition and suggest that competitive sialyltransferase inhibition might offer new therapeutic opportunities against skin fibrosis.


Subject(s)
Cell Differentiation , Cell Proliferation , Fibroblasts , Myofibroblasts , Sialic Acids , Skin , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Skin/metabolism , Skin/pathology , Sialic Acids/metabolism , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/drug effects , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Sialyltransferases/metabolism , Sialyltransferases/genetics , Signal Transduction/drug effects , Cells, Cultured
6.
Int Immunopharmacol ; 136: 112338, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38850787

ABSTRACT

Cardiac fibrosis is a typical feature of cardiac pathological remodeling, which is associated with adverse clinical outcomes and has no effective therapy. Nicotine is an important risk factor for cardiac fibrosis, yet its underlying molecular mechanism remains poorly understood. This study aimed to identify its potential molecular mechanism in nicotine-induced cardiac fibrosis. Our results showed nicotine exposure led to the proliferation and transformation of cardiac fibroblasts (CFs) into myofibroblasts (MFs) by impairing autophagy flux. Through the use of drug affinity responsive target stability (DARTS) assay, cellular thermal shift assay (CETSA), and surface plasmon resonance (SPR) technology, it was discovered that nicotine directly increased the stability and protein levels of lactate dehydrogenase A (LDHA) by binding to it. Nicotine treatment impaired autophagy flux by regulating the AMPK/mTOR signaling pathway, impeding the nuclear translocation of transcription factor EB (TFEB), and reducing the activity of cathepsin B (CTSB). In vivo, nicotine treatment exacerbated cardiac fibrosis induced in spontaneously hypertensive rats (SHR) and worsened cardiac function. Interestingly, the absence of LDHA reversed these effects both in vitro and in vivo. Our study identified LDHA as a novel nicotine-binding protein that plays a crucial role in mediating cardiac fibrosis by blocking autophagy flux. The findings suggest that LDHA could potentially serve as a promising target for the treatment of cardiac fibrosis.


Subject(s)
Autophagy , Fibrosis , Nicotine , Animals , Autophagy/drug effects , Rats , Male , Rats, Inbred SHR , Signal Transduction/drug effects , Myocardium/pathology , Myocardium/metabolism , Lactate Dehydrogenase 5/metabolism , Cells, Cultured , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , TOR Serine-Threonine Kinases/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Rats, Sprague-Dawley
7.
J Control Release ; 370: 421-437, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701884

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with a high mortality rate due to limited treatment options. Current therapies cannot effectively reverse the damage caused by IPF. Research suggests that promoting programmed cell death (apoptosis) in myofibroblasts, the key cells driving fibrosis, could be a promising strategy. However, inducing apoptosis in healthy cells like epithelial and endothelial cells can cause unwanted side effects. This project addresses this challenge by developing a targeted approach to induce apoptosis specifically in myofibroblasts. We designed liposomes (LPS) decorated with peptides that recognize VCAM-1, a protein highly expressed on myofibroblasts in fibrotic lungs. These VCAM1-targeted LPS encapsulate Venetoclax (VNT), a small molecule drug that inhibits BCL-2, an anti-apoptotic protein. By delivering VNT directly to myofibroblasts, we hypothesize that VCAM1-VNT-LPS can selectively induce apoptosis in these cells, leading to reduced fibrosis and improved lung function. We successfully characterized VCAM1-VNT-LPS for size, surface charge, and drug loading efficiency. Additionally, we evaluated their stability over three months at different temperatures. In vitro and in vivo studies using a bleomycin-induced mouse model of lung fibrosis demonstrated the therapeutic potential of VCAM1-VNT-LPS. These studies showed a reduction in fibrosis-associated proteins (collagen, α-SMA, VCAM1) and BCL-2, while simultaneously increasing apoptosis in myofibroblasts. These findings suggest that VCAM1-targeted delivery of BCL-2 inhibitors using liposomes presents a promising and potentially selective therapeutic approach for IPF.


Subject(s)
Apoptosis , Bridged Bicyclo Compounds, Heterocyclic , Liposomes , Mice, Inbred C57BL , Nanoparticles , Proto-Oncogene Proteins c-bcl-2 , Sulfonamides , Vascular Cell Adhesion Molecule-1 , Animals , Proto-Oncogene Proteins c-bcl-2/metabolism , Nanoparticles/administration & dosage , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Vascular Cell Adhesion Molecule-1/metabolism , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Apoptosis/drug effects , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Humans , Lung/pathology , Lung/drug effects , Lung/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Male , Mice , Bleomycin/administration & dosage
8.
Arch Oral Biol ; 163: 105980, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38692246

ABSTRACT

OBJECTIVE: To determine the effect of hyaluronic acid (HA) degradation by hyaluronidase (HYAL) in inhibiting collagen fiber production by rat periodontal ligament cells (rPDLCs). DESIGN: Primary rPDLCs were isolated from the euthanized rats and used for in vitro experiments. The appropriate HYAL concentration was determined through CCK-8 testing for cytotoxicity detection and Alizarin red staining for mineralization detection. RT-qPCR and western blot assays were conducted to assess the effect of HYAL, with or without TGF-ß, on generation of collagen fiber constituents and expression of actin alpha 2, smooth muscle (ACTA2) of rPDLCs. RESULTS: Neither cell proliferation nor mineralization were significantly affected by treatment with 4 U/mL HYAL. HYAL (4 U/mL) alone downregulated type I collagen fiber (Col1a1 and Col1a2) and Acta2 mRNA expression; however, ACTA2 and COL1 protein levels were only downregulated by HYAL treatment after TGF-ß induction. CONCLUSIONS: Treatment of rPDLCs with HYAL can inhibit TGF-ß-induced collagen matrix formation and myofibroblast transformation.


Subject(s)
Cell Proliferation , Collagen , Fibroblasts , Hyaluronoglucosaminidase , Myofibroblasts , Periodontal Ligament , Transforming Growth Factor beta , Animals , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Hyaluronoglucosaminidase/pharmacology , Rats , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Transforming Growth Factor beta/metabolism , Collagen/metabolism , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Hyaluronic Acid/pharmacology , Cells, Cultured , Rats, Sprague-Dawley , Actins/metabolism , Blotting, Western , In Vitro Techniques , Collagen Type I/metabolism , Biomarkers/metabolism , Real-Time Polymerase Chain Reaction , Male , RNA, Messenger/metabolism
9.
Sci Rep ; 14(1): 12111, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802470

ABSTRACT

Alkaline burns to the cornea lead to loss of corneal transparency, which is essential for normal vision. We used a rat corneal alkaline burn model to investigate the effect of ophthalmic trimebutine solution on healing wounds caused by alkaline burns. Trimebutine, an inhibitor of the high-mobility group box 1-receptor for advanced glycation end products, when topically applied to the burned cornea, suppressed macrophage infiltration in the early phase and neutrophil infiltration in the late phase at the wound site. It also inhibited neovascularization and myofibroblast development in the late phase. Furthermore, trimebutine effectively inhibited interleukin-1ß expression in the injured cornea. It reduced scar formation by decreasing the expression of type III collagen. These findings suggest that trimebutine may represent a novel therapeutic strategy for corneal wounds, not only through its anti-inflammatory effects but also by preventing neovascularization.


Subject(s)
Alkalies , Burns, Chemical , Cornea , Disease Models, Animal , Eye Burns , Wound Healing , Animals , Burns, Chemical/drug therapy , Burns, Chemical/pathology , Burns, Chemical/metabolism , Rats , Eye Burns/chemically induced , Eye Burns/drug therapy , Eye Burns/pathology , Alkalies/adverse effects , Cornea/metabolism , Cornea/pathology , Cornea/drug effects , Wound Healing/drug effects , Interleukin-1beta/metabolism , Male , Macrophages/drug effects , Macrophages/metabolism , Corneal Injuries/drug therapy , Corneal Injuries/metabolism , Corneal Injuries/pathology , Corneal Injuries/chemically induced , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Rats, Sprague-Dawley , Collagen Type III/metabolism , Receptor for Advanced Glycation End Products/metabolism , Anti-Inflammatory Agents/pharmacology , Ophthalmic Solutions , Myofibroblasts/metabolism , Myofibroblasts/drug effects
10.
Sci Rep ; 14(1): 9976, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38693148

ABSTRACT

Inflammation and fibrosis often occur in the kidney after acute injury, resulting in chronic kidney disease and consequent renal failure. Recent studies have indicated that lymphangiogenesis can drive renal inflammation and fibrosis in injured kidneys. However, whether and how this pathogenesis affects the contralateral kidney remain largely unknown. In our study, we uncovered a mechanism by which the contralateral kidney responded to injury. We found that the activation of mineralocorticoid receptors and the increase in vascular endothelial growth factor C in the contralateral kidney after unilateral ureteral obstruction could promote lymphangiogenesis. Furthermore, mineralocorticoid receptor activation in lymphatic endothelial cells resulted in the secretion of myofibroblast markers, thereby contributing to renal fibrosis. We observed that this process could be attenuated by administering the mineralocorticoid receptor blocker eplerenone, which, prevented the development of fibrotic injury in the contralateral kidneys of rats with unilateral ureteral obstruction. These findings offer valuable insights into the intricate mechanisms underlying kidney injury and may have implications for the development of therapeutic strategies to mitigate renal fibrosis in the context of kidney disease.


Subject(s)
Eplerenone , Fibrosis , Kidney , Lymphangiogenesis , Mineralocorticoid Receptor Antagonists , Ureteral Obstruction , Animals , Eplerenone/pharmacology , Lymphangiogenesis/drug effects , Rats , Fibrosis/drug therapy , Kidney/metabolism , Kidney/drug effects , Kidney/pathology , Ureteral Obstruction/drug therapy , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Ureteral Obstruction/complications , Mineralocorticoid Receptor Antagonists/pharmacology , Male , Receptors, Mineralocorticoid/metabolism , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , Vascular Endothelial Growth Factor C/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Rats, Sprague-Dawley , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Myofibroblasts/pathology
11.
PLoS One ; 19(5): e0285655, 2024.
Article in English | MEDLINE | ID: mdl-38753593

ABSTRACT

BACKGROUND: Chronic rhinosinusitis (CRS) is an inflammatory disease affecting the sinuses or nose. Persistent inflammatory responses can lead to tissue remodeling, which is a pathological characteristics of CRS. Activation of fibroblasts in the nasal mucosal stroma, differentiation and collagen deposition, and subepithelial fibrosis have been associated with CRS. OBJECTIVES: We aimed to assess the inhibitory effects of doxycycline and deoxycholic acid-polyethyleneimine conjugate (DA3-Doxy) on myofibroblast differentiation and extracellular matrix (ECM) production in nasal fibroblasts stimulated with TGF-ß1. METHODS: To enhance efficacy, we prepared DA3-Doxy using a conjugate of low-molecular-weight polyethyleneimine (PEI) (MW 1800) and deoxycholic acid (DA) and Doxy. The synthesis of the DA3-Doxy polymer was confirmed using nuclear magnetic resonance, and the critical micelle concentration required for cationic micelle formation through self-assembly was determined. Subsequently, the Doxy loading efficiency of DA3 was assessed. The cytotoxicity of Doxy, DA3, PEI, and DA-Doxy in nasal fibroblasts was evaluated using the WST-1 assay. The anti-tissue remodeling and anti-inflammatory effects of DA3-Doxy and DA3 were examined using real-time polymerase chain reaction (Real-time PCR), immunocytochemistry, western blot, and Sircol assay. RESULTS: Both DA3 and DA3-Doxy exhibited cytotoxicity at 10 µg/ml in nasal fibroblasts. Doxy partially inhibited α-smooth muscle actin, collagen types I and III, and fibronectin. However, DA3-Doxy significantly inhibited α-SMA, collagen types I and III, and fibronectin at 5 µg/ml. DA3-Doxy also modulated TGF-ß1-induced changes in the expression of MMP 1, 2, and 9. Nonetheless, TGF-ß1-induced expression of MMP3 was further increased by DA3-Doxy. The expression of TIMP 1 and 2 was partially reduced with 5 µg/ml DA3-Doxy. CONCLUSIONS: Although initially developed for the delivery of genetic materials or drugs, DA3 exhibits inhibitory effects on myofibroblast differentiation and ECM production. Therefore, it holds therapeutic potential for CRS, and a synergistic effect can be expected when loaded with CRS treatment drugs.


Subject(s)
Cell Differentiation , Deoxycholic Acid , Doxycycline , Fibroblasts , Polyethyleneimine , Humans , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Deoxycholic Acid/chemistry , Deoxycholic Acid/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Cell Differentiation/drug effects , Doxycycline/pharmacology , Doxycycline/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Transforming Growth Factor beta1/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Nasal Mucosa/drug effects , Nasal Mucosa/metabolism , Nasal Mucosa/cytology , Actins/metabolism
12.
Int Heart J ; 65(3): 487-497, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38749755

ABSTRACT

Myocardial fibrosis is a pathological feature of doxorubicin-induced chronic cardiotoxicity that severely affects the prognosis of oncology patients. However, the specific cellular and molecular mediators driving doxorubicin-induced cardiac fibrosis, and the relative impact of different cell populations on cardiac fibrosis, remain unclear.This study aimed to explore the mechanism of doxorubicin-induced cardiotoxicity and myocardial fibrosis and to find potential therapeutic targets. Single-cell RNA sequencing was used to analyze the transcriptome of non-cardiomyocytes from normal and doxorubicin-induced chronic cardiotoxicity in mouse model heart tissue.We established a mouse model of doxorubicin-induced cardiotoxicity with a well-defined fibrotic phenotype. Analysis of single-cell sequencing results showed that fibroblasts were the major origin of extracellular matrix in doxorubicin-induced myocardial fibrosis. Further resolution of fibroblast subclusters showed that resting fibroblasts were converted to matrifibrocytes and then to myofibroblasts to participate in the myocardial remodeling process in response to doxorubicin treatment. Ctsb expression was significantly upregulated in fibroblasts after doxorubicin-induced.This study provides a comprehensive map of the non-cardiomyocyte landscape at high resolution, reveals multiple cell populations contributing to pathological remodeling of the cardiac extracellular matrix, and identifies major cellular sources of myofibroblasts and dynamic gene-expression changes in fibroblast activation. Finally, we used this strategy to detect potential therapeutic targets and identified Ctsb as a specific target for fibroblasts in doxorubicin-induced myocardial fibrosis.


Subject(s)
Cardiotoxicity , Doxorubicin , Fibrosis , Single-Cell Analysis , Doxorubicin/adverse effects , Animals , Mice , Single-Cell Analysis/methods , Myocardium/pathology , Myocardium/metabolism , Antibiotics, Antineoplastic/toxicity , Antibiotics, Antineoplastic/adverse effects , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling/methods , Transcriptome , Male , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Mice, Inbred C57BL
13.
Cell Mol Gastroenterol Hepatol ; 18(1): 105-131, 2024.
Article in English | MEDLINE | ID: mdl-38614455

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel disease is associated with carcinogenesis, which limits the prognosis of the patients. The local expression of proteinases and proteinase-activated receptor 1 (PAR1) increases in inflammatory bowel disease. The present study investigated the therapeutic effects of PAR1 antagonism on colitis-associated carcinogenesis. METHODS: A colitis-associated carcinogenesis model was prepared in mice by treatment with azoxymethane (AOM) and dextran sulfate sodium (DSS). PAR1 antagonist E5555 was administered in long- and short-term protocol, starting on the day of AOM injection and 1 week after completing AOM/DSS treatment, respectively. The fecal samples were collected for metagenome analysis of gut microbiota. The intestinal myofibroblasts of the Crohn's disease patients were used to elucidate underlying cellular mechanisms. Caco-2 cells were used to investigate a possible source of PAR1 agonist proteinases. RESULTS: AOM/DSS model showed weight loss, diarrhea, tumor development, inflammation, fibrosis, and increased production of inflammatory cytokines. The ß-diversity, but not α-diversity, of microbiota significantly differed between AOM/DSS and control mice. E5555 alleviated these pathological changes and altered the microbiota ß-diversity in AOM/DSS mice. The thrombin expression was up-regulated in tumor and non-tumor areas, whereas PAR1 mRNA expression was higher in tumor areas compared with non-tumor areas. E5555 inhibited thrombin-triggered elevation of cytosolic Ca2+ concentration and ERK1/2 phosphorylation, as well as IL6-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation in intestinal myofibroblasts. Caco-2 cell-conditioned medium contained immunoreactive thrombin, which cleaved the recombinant protein containing the extracellular domain of PAR1 at the thrombin cleavage site. CONCLUSIONS: PAR1 antagonism is proposed to be a novel therapeutic strategy for treatment of inflammatory bowel disease and its associated carcinogenesis.


Subject(s)
Azoxymethane , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Receptor, PAR-1 , Animals , Receptor, PAR-1/metabolism , Receptor, PAR-1/antagonists & inhibitors , Humans , Mice , Caco-2 Cells , Dextran Sulfate/toxicity , Azoxymethane/toxicity , Gastrointestinal Microbiome/drug effects , Male , Colitis/complications , Colitis/chemically induced , Colitis/pathology , Colitis/drug therapy , Carcinogenesis/drug effects , Carcinogenesis/pathology , STAT3 Transcription Factor/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Myofibroblasts/drug effects , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/immunology , Thrombin/metabolism , Mice, Inbred C57BL , Crohn Disease/pathology , Crohn Disease/drug therapy , Crohn Disease/microbiology , Crohn Disease/chemically induced
14.
Lung ; 202(3): 343-356, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678499

ABSTRACT

BACKGROUND: Severe asthma, characterized by inflammation and airway remodeling, involves fibroblast differentiation into myofibroblasts expressing α-SMA. This process leads to the production of fibronectin and connective tissue growth factor (CTGF), driven by factors such as transforming growth factor (TGF)-ß. Furthermore, the persistent presence of myofibroblasts is associated with resistance to apoptosis and mitochondrial dysfunction. The chemokine (C-X3-C motif) ligand 1 (CX3CL1) plays a role in tissue fibrosis. However, it is currently unknown whether neutralization of CX3CL1 decreases TGF-ß-induced fibroblast differentiation and mitochondrial dysfunction in normal human lung fibroblasts (NHLFs). METHODS: CX3CL1/C-X3-C motif chemokine receptor 1 (CX3CR1), CX3CL1 was analyzed by immunofluorescence (IF) or immunohistochemical (IHC) staining of ovalbumin-challenged mice. CX3CL1 release was detected by ELISA. TGF-ß-induced CTGF, fibronectin, and α-SMA expression were evaluated in NHLFs following neutralization of CX3CL1 (TP213) treatment for the indicated times by Western blotting or IF staining. Mitochondrion function was detected by a JC-1 assay and seahorse assay. Cell apoptosis was observed by a terminal uridine nick-end labeling (TUNEL) assay. RESULTS: An increase in CX3CL1 expression was observed in lung tissues from mice with ovalbumin-induced asthma by IF staining. CX3CR1 was increased in the subepithelial layer of the airway by IHC staining. Moreover, CX3CR1 small interfering (si)RNA downregulated TGF-ß-induced CTGF and fibronectin expression in NHLFs. CX3CL1 induced CTGF and fibronectin expression in NHLFs. TGF-ß-induced CX3CL1 secretion from NHLFs. Furthermore, TP213 decreased TGF-ß-induced CTGF, fibronectin, and α-SMA expression in NHLFs. Mitochondrion-related differentially expressed genes (DEGs) were examined after CX3CL1 neutralization in TGF-ß-treated NHLFs. TP213 alleviated TGF-ß-induced mitochondrial dysfunction and apoptosis resistance in NHLFs. CX3CL1 induced p65, IκBα, and IKKα phosphorylation in a time-dependent manner. Furthermore, CX3CL1-induced fibronectin expression and JC-1 monomer were decreased by p65 siRNA. TP213 reduced TGF-ß-induced p65 and α-SMA expression in NHLFs. CONCLUSIONS: These findings suggest that neutralizing CX3CL1 attenuates lung fibroblast activation and mitochondrial dysfunction. Understanding the impacts of CX3CL1 neutralization on fibroblast mitochondrial function could contribute to the development of therapeutic strategies for managing airway remodeling in severe asthma.


Subject(s)
Apoptosis , CX3C Chemokine Receptor 1 , Cell Differentiation , Chemokine CX3CL1 , Connective Tissue Growth Factor , Fibroblasts , Fibronectins , Mitochondria , Pulmonary Fibrosis , Transforming Growth Factor beta , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/genetics , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Humans , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Cell Differentiation/drug effects , Apoptosis/drug effects , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Transforming Growth Factor beta/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Fibronectins/metabolism , Mice , Actins/metabolism , Lung/pathology , Lung/metabolism , NF-kappa B/metabolism , Signal Transduction , Asthma/metabolism , Asthma/pathology , Disease Models, Animal , Cells, Cultured , Myofibroblasts/metabolism , Myofibroblasts/pathology , Myofibroblasts/drug effects , Ovalbumin
15.
Phytomedicine ; 129: 155620, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38669964

ABSTRACT

BACKGROUND: Activation of myofibroblasts, linked to oxidative stress, emerges as a pivotal role in the progression of pulmonary fibrosis (PF). Our prior research has underscored the therapeutic promise of tanshinone IIA (Tan-IIA) in mitigating PF by enhancing nuclear factor-erythroid 2-related factor 2 (Nrf2) activity. Nevertheless, the molecular basis through which Tan-IIA influences Nrf2 activity has yet to be fully elucidated. METHODS: The influence of Tan-IIA on PF was assessed in vivo and in vitro models. Inhibitors, overexpression plasmids, and small interfering RNA (siRNA) were utilized to probe its underlying mechanism of action in vitro. RESULTS: We demonstrate that Tan-IIA effectively activates the kelch-like ECH-associated protein 1 (Keap1)-Nrf2 antioxidant pathway, which in turn inhibits myofibroblast activation and ameliorates PF. Notably, the stability and nucleo-cytoplasmic shuttling of Nrf2 is shown to be dependent on augmented autophagic flux, which is in alignment with the observation that Tan-IIA induces autophagy. Inhibition of autophagy, conversely, fosters the activation of extracellular matrix (ECM)-producing myofibroblasts. Further, Tan-IIA initiates an autophagy program through the sestrin 2 (Sesn2)-sequestosome 1 (Sqstm1) signaling axis, crucial for protecting Nrf2 from Keap1-mediated degradation. Meanwhile, these findings were corroborated in a murine model of PF. CONCLUSION: Collectively, we observed for the first time that the Sqstm1-Sesn2 axis-mediated autophagic degradation of Keap1 effectively prevents myofibroblast activation and reduces the synthesis of ECM. This autophagy-dependent degradation of Keap1 can be initiated by the Tan-IIA treatment, which solidifies its potential as an Nrf2-modulating agent for PF treatment.


Subject(s)
Abietanes , Autophagy , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Pulmonary Fibrosis , Sequestosome-1 Protein , Signal Transduction , Animals , Humans , Male , Mice , Abietanes/pharmacology , Autophagy/drug effects , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Sequestosome-1 Protein/metabolism , Sestrins , Signal Transduction/drug effects
16.
World J Gastroenterol ; 30(15): 2143-2154, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38681990

ABSTRACT

BACKGROUND: Liver fibrosis is a compensatory response during the tissue repair process in chronic liver injury, and finally leads to liver cirrhosis or even hepatocellular carcinoma. The pathogenesis of hepatic fibrosis is associated with the progressive accumulation of activated hepatic stellate cells (HSCs), which can transdifferentiate into myofibroblasts to produce an excess of the extracellular matrix (ECM). Myofibroblasts are the main source of the excessive ECM responsible for hepatic fibrosis. Therefore, activated hepatic stellate cells (aHSCs), the principal ECM producing cells in the injured liver, are a promising therapeutic target for the treatment of hepatic fibrosis. AIM: To explore the effect of taurine on aHSC proliferation and the mechanisms involved. METHODS: Human HSCs (LX-2) were randomly divided into five groups: Normal control group, platelet-derived growth factor-BB (PDGF-BB) (20 ng/mL) treated group, and low, medium, and high dosage of taurine (10 mmol/L, 50 mmol/L, and 100 mmol/L, respectively) with PDGF-BB (20 ng/mL) treated group. Cell Counting Kit-8 method was performed to evaluate the effect of taurine on the viability of aHSCs. Enzyme-linked immunosorbent assay was used to estimate the effect of taurine on the levels of reactive oxygen species (ROS), malondialdehyde, glutathione, and iron concentration. Transmission electron microscopy was applied to observe the effect of taurine on the autophagosomes and ferroptosis features in aHSCs. Quantitative real-time polymerase chain reaction and Western blot analysis were performed to detect the effect of taurine on the expression of α-SMA, Collagen I, Fibronectin 1, LC3B, ATG5, Beclin 1, PTGS2, SLC7A11, and p62. RESULTS: Taurine promoted the death of aHSCs and reduced the deposition of the ECM. Treatment with taurine could alleviate autophagy in HSCs to inhibit their activation, by decreasing autophagosome formation, downregulating LC3B and Beclin 1 protein expression, and upregulating p62 protein expression. Meanwhile, treatment with taurine triggered ferroptosis and ferritinophagy to eliminate aHSCs characterized by iron overload, lipid ROS accumulation, glutathione depletion, and lipid peroxidation. Furthermore, bioinformatics analysis demonstrated that taurine had a direct targeting effect on nuclear receptor coactivator 4, exhibiting the best average binding affinity of -20.99 kcal/mol. CONCLUSION: Taurine exerts therapeutic effects on liver fibrosis via mechanisms that involve inhibition of autophagy and trigger of ferroptosis and ferritinophagy in HSCs to eliminate aHSCs.


Subject(s)
Autophagy , Cell Proliferation , Ferroptosis , Hepatic Stellate Cells , Liver Cirrhosis , Reactive Oxygen Species , Taurine , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Autophagy/drug effects , Taurine/pharmacology , Ferroptosis/drug effects , Liver Cirrhosis/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Cell Proliferation/drug effects , Reactive Oxygen Species/metabolism , Becaplermin/pharmacology , Becaplermin/metabolism , Cell Line , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Cell Survival/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Signal Transduction/drug effects
17.
Ren Fail ; 46(1): 2327498, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38666363

ABSTRACT

Iguratimod is a novel synthetic, small-molecule immunosuppressive agent used to treat rheumatoid arthritis. Through ongoing exploration of its role and mechanisms of action, iguratimod has been observed to have antifibrotic effects in the lung and skin; however, its effect on renal fibrosis remains unknown. This study aimed to investigate whether iguratimod could affect renal fibrosis progression. Three different concentrations of iguratimod (30 mg/kg/day, 10 mg/kg/day, and 3 mg/kg/day) were used to intervene in unilateral ureteral obstruction (UUO) model mice. Iguratimod at 10 mg/kg/day was observed to be effective in slowing UUO-mediated renal fibrosis. In addition, stimulating bone marrow-derived macrophages with IL-4 and/or iguratimod, or with TGF-ß and iguratimod or SRC inhibitors in vitro, suggested that iguratimod mitigates the progression of renal fibrosis in UUO mice, at least in part, by inhibiting the IL-4/STAT6 signaling pathway to attenuate renal M2 macrophage infiltration, as well as by impeding SRC activation to reduce macrophage-myofibroblast transition. These findings reveal the potential of iguratimod as a treatment for renal disease.


Subject(s)
Disease Models, Animal , Fibrosis , Interleukin-4 , Macrophages , STAT6 Transcription Factor , Sulfonamides , Ureteral Obstruction , Animals , Ureteral Obstruction/complications , Mice , Macrophages/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Interleukin-4/metabolism , STAT6 Transcription Factor/metabolism , Male , Myofibroblasts/drug effects , Chromones/pharmacology , Chromones/therapeutic use , Kidney/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Kidney Diseases/pathology , Kidney Diseases/drug therapy , Mice, Inbred C57BL , Immunosuppressive Agents/pharmacology
18.
Int Immunopharmacol ; 132: 111999, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38581994

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a heterogeneous group of lung diseases with different etiologies and characterized by progressive fibrosis. This disease usually causes pulmonary structural remodeling and decreased pulmonary function. The median survival of IPF patients is 2-5 years. Predominantly accumulation of type II innate immune cells accelerates fibrosis progression by secreting multiple pro-fibrotic cytokines. Group 2 innate lymphoid cells (ILC2) and monocytes/macrophages play key roles in innate immunity and aggravate the formation of pro-fibrotic environment. As a potent immunosuppressant, tacrolimus has shown efficacy in alleviating the progression of pulmonary fibrosis. In this study, we found that tacrolimus is capable of suppressing ILC2 activation, monocyte differentiation and the interaction of these two cells. This effect further reduced activation of monocyte-derived macrophages (Mo-M), thus resulting in a decline of myofibroblast activation and collagen deposition. The combination of tacrolimus and nintedanib was more effective than either drug alone. This study will reveal the specific process of tacrolimus alleviating pulmonary fibrosis by regulating type II immunity, and explore the potential feasibility of tacrolimus combined with nintedanib in the treatment of pulmonary fibrosis. This project will provide new ideas for clinical optimization of anti-pulmonary fibrosis drug strategies.


Subject(s)
Idiopathic Pulmonary Fibrosis , Immunosuppressive Agents , Mice, Inbred C57BL , Monocytes , Tacrolimus , Tacrolimus/therapeutic use , Tacrolimus/pharmacology , Animals , Monocytes/drug effects , Monocytes/immunology , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Mice , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/pharmacology , Humans , Lymphocytes/drug effects , Lymphocytes/immunology , Immunity, Innate/drug effects , Indoles/therapeutic use , Indoles/pharmacology , Macrophages/drug effects , Macrophages/immunology , Disease Progression , Lung/pathology , Lung/drug effects , Lung/immunology , Cells, Cultured , Male , Cytokines/metabolism , Myofibroblasts/drug effects , Cell Differentiation/drug effects , Disease Models, Animal
19.
Exp Eye Res ; 242: 109884, 2024 May.
Article in English | MEDLINE | ID: mdl-38570181

ABSTRACT

Recent studies in rabbits and case reports in humans have demonstrated the efficacy of topical losartan in the treatment of corneal scarring fibrosis after a wide range of injuries, including chemical burns, infections, surgical complications, and some diseases. It is hypothesized that the effect of losartan on the fibrotic corneal stroma occurs through a two-phase process in which losartan first triggers the elimination of myofibroblasts by directing their apoptosis via inhibition of extracellular signal-regulated kinase (ERK)-mediated signal transduction, and possibly through signaling effects on the viability and development of corneal fibroblast and fibrocyte myofibroblast precursor cells. This first step likely occurs within a week or two in most corneas with fibrosis treated with topical losartan, but the medication must be continued for much longer until the epithelial basement membrane (EBM) is fully regenerated or new myofibroblasts will develop from precursor cells. Once the myofibroblasts are eliminated from the fibrotic stroma, corneal fibroblasts can migrate into the fibrotic tissue and reabsorb/reorganize the disordered extracellular matrix (ECM) previously produced by the myofibroblasts. This second stage is longer and more variable in different eyes of rabbits and humans, and accounts for most of the variability in the time it takes for the stromal opacity to be markedly reduced by topical losartan treatment. Eventually, keratocytes reemerge in the previously fibrotic stromal tissue to fine-tune the collagens and other ECM components and maintain the normal structure of the corneal stroma. The efficacy of losartan in the prevention and treatment of corneal fibrosis suggests that it acts as a surrogate for the EBM, by suppressing TGF beta-directed scarring of the wounded corneal stroma, until control over TGF beta action is re-established by a healed EBM, while also supporting regeneration of the EBM by allowing corneal fibroblasts to occupy the subepithelial stroma in the place of myofibroblasts.


Subject(s)
Corneal Stroma , Fibrosis , Losartan , Myofibroblasts , Losartan/therapeutic use , Corneal Stroma/drug effects , Corneal Stroma/metabolism , Corneal Stroma/pathology , Fibrosis/drug therapy , Humans , Animals , Myofibroblasts/pathology , Myofibroblasts/drug effects , Rabbits , Corneal Diseases/drug therapy , Corneal Diseases/pathology , Angiotensin II Type 1 Receptor Blockers , Administration, Topical
20.
Ecotoxicol Environ Saf ; 276: 116309, 2024 May.
Article in English | MEDLINE | ID: mdl-38599156

ABSTRACT

Emerging evidence has suggested that exposure to PM2.5 is a significant contributing factor to the development of chronic obstructive pulmonary disease (COPD). However, the underlying biological effects and mechanisms of PM2.5 in COPD pathology remain elusive. In this study, we aimed to investigate the implication and regulatory effect of biomass fuels related-PM2.5 (BRPM2.5) concerning the pathological process of fibroblast-to-myofibroblast transition (FMT) in the context of COPD. In vivo experimentation revealed that exposure to biofuel smoke was associated with airway inflammation in rats. After 4 weeks of exposure, there was inflammation in the small airways, but no significant structural changes in the airway walls. However, after 24 weeks, airway remodeling occurred due to increased collagen deposition, myofibroblast proliferation, and tracheal wall thickness. In vitro, cellular immunofluorescence results showed that with stimulation of BRPM2.5 for 72 h, the cell morphology of fibroblasts changed significantly, most of the cells changed from spindle-shaped to star-shaped irregular, α-SMA stress fibers appeared in the cytoplasm and the synthesis of type I collagen increased. The collagen gel contraction experiment showed that the contractility of fibroblasts was enhanced. The expression level of TRPC1 in fibroblasts was increased. Specific siRNA-TRPC1 blocked BRPM2.5-induced FMT and reduced cell contractility. Additionally, specific siRNA-TRPC1 resulted in a decrease in the augment of intracellular Ca2+ concentration ([Ca2+]i) induced by BRPM2.5. Notably, it was found that the PI3K inhibitor, LY294002, inhibited enhancement of AKT phosphorylation level, FMT occurrence, and elevation of TRPC1 protein expression induced by BRPM2.5. The findings indicated that BRPM2.5 is capable of inducing the FMT, with the possibility of mediation by PI3K/AKT/TRPC1. These results hold potential implications for the understanding of the molecular mechanisms involved in BRPM2.5-induced COPD and may aid in the development of novel therapeutic strategies for pathological conditions characterized by fibrosis.


Subject(s)
Fibroblasts , Lung , Myofibroblasts , Particulate Matter , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , TRPC Cation Channels , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Fibroblasts/drug effects , Rats , Myofibroblasts/drug effects , Particulate Matter/toxicity , Lung/drug effects , Lung/pathology , TRPC Cation Channels/metabolism , Male , Biomass , Signal Transduction/drug effects , Rats, Sprague-Dawley , Pulmonary Disease, Chronic Obstructive/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...