Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.120
Filter
1.
Respir Res ; 25(1): 282, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014439

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterized by chronic bronchitis, emphysema and vascular remodelling. The disease is associated with hypoxia, inflammation and oxidative stress. Lung fibroblasts are important cells in remodelling processes in COPD, as main producers of extracellular matrix proteins but also in synthesis of growth factors and inflammatory mediators. METHODS: In this study we aimed to investigate if there are differences in how primary distal lung fibroblasts obtained from COPD patients and healthy subjects respond to hypoxia (1% O2) and pro-fibrotic stimuli with TGF-ß1 (10 ng/mL). Genes and proteins associated with oxidative stress, endoplasmic reticulum stress, remodelling and inflammation were analysed with RT-qPCR and ELISA. RESULTS: Hypoxia induced differences in expression of genes involved in oxidative stress (SOD3 and HIF-1α), ER stress (IRE1, PARK and ATF6), apoptosis (c-Jun and Bcl2) and remodelling (5HTR2B, Collagen7 and VEGFR2) in lung fibroblasts from COPD subjects compared to control subjects, where COPD fibroblasts were in general less responsive. The release of VEGF-C was increased after hypoxia, whereas TGF-ß significantly reduced the VEGF response to hypoxia and the release of HGF. COPD fibroblasts had a higher release of IL-6, IL-8, MCP-1 and PGE2 compared to lung fibroblasts from control subjects. The release of inflammatory mediators was less affected by hypoxia, whereas TGFß1 induced differences in inflammatory profile between fibroblasts from COPD and control subjects. CONCLUSION: These results suggest that there is an alteration of gene regulation of various stress responses and remodelling associated mediator release that is related to COPD and hypoxia, where fibroblasts from COPD patients have a deficient response.


Subject(s)
Fibroblasts , Lung , Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Male , Female , Middle Aged , Cells, Cultured , Aged , Lung/metabolism , Lung/pathology , Cell Hypoxia/physiology , Oxidative Stress/physiology , Inflammation Mediators/metabolism , Endoplasmic Reticulum Stress/physiology , Inflammation/metabolism , Inflammation/pathology , Hypoxia/metabolism , Transforming Growth Factor beta1/metabolism , Case-Control Studies
2.
Mol Biol Rep ; 51(1): 812, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007943

ABSTRACT

BACKGROUND: Estrogen has a protective impact on acute kidney injury (AKI); moreover, reducing the daily intake of calories impedes developing diseases. The present study aimed to determine the effects of calorie restriction (CR) and time restriction (TR) diets on the expression of silent information regulator 2 homolog 1 (SIRT1), transforming growth factor beta 1 (TGF-ß1), and other indicators in the presence and absence of ovaries in AKI female rats. METHODS: The female rats were divided into two groups, ovariectomized (OVX) and sham, and were placed on CR and TR diets for eight weeks; afterward, AKI was induced by injecting glycerol, and kidney injury indicators and biochemical parameters were measured before and after AKI. RESULTS: After AKI, the levels of urine albumin excretion rate, urea, and creatinine in serum, and TGF-ß1 increased, while creatinine clearance and SIRT1 decreased in kidney tissue. CR improved kidney indicators and caused a reduction in TGF-ß1 and an increase in SIRT1 in ovary-intact rats. Moreover, CR prevented total antioxidant capacity (TAC) decrease and malondialdehyde (MDA) increase resulting from AKI. Before AKI, an increase in body weight, fasting blood sugar (FBS), low-density lipoprotein (LDL), triglyceride (TG), and total cholesterol (TC), and a decrease in high-density lipoprotein (HDL) were observed in OVX rats compared to sham rats, but CR prevented these changes. The effects of TR were similar to those of CR in all indicators except for TGF-ß1, SIRT1, urea, creatinine, and albumin. CONCLUSION: The present study indicated that CR is more effective than TR in preventing AKI, probably by increasing SIRT1 and decreasing TGF-ß1 in ovary-intact animals.


Subject(s)
Acute Kidney Injury , Caloric Restriction , Sirtuin 1 , Transforming Growth Factor beta1 , Animals , Female , Sirtuin 1/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Acute Kidney Injury/metabolism , Rats , Caloric Restriction/methods , Kidney/metabolism , Kidney/pathology , Menopause/metabolism , Ovariectomy , Creatinine/blood , Disease Models, Animal , Body Weight
3.
PLoS One ; 19(7): e0305927, 2024.
Article in English | MEDLINE | ID: mdl-39024326

ABSTRACT

Fibrotic skin diseases, such as keloids, are pathological results of aberrant tissue healing and are characterized by overgrowth of dermal fibroblasts. Remdesivir (RD), an antiviral drug, has been reported to have pharmacological activities in a wide range of fibrotic diseases. However, whether RD function on skin fibrosis remains unclear. Therefore, in our study, we explored the potential effect and mechanisms of RD on skin fibrosis both in vivo and in vitro. As expected, the results demonstrated that RD alleviated BLM-induced skin fibrosis and attenuates the gross weight of keloid tissues in vivo. Further studies suggested that RD suppressed fibroblast activation and autophagy both in vivo and in vitro. In addition, mechanistic research showed that RD attenuated fibroblasts activation by the TGF-ß1/Smad signaling pathway and inhibited fibroblasts autophagy by the PI3K/Akt/mTOR signaling pathway. In summary, our results demonstrate therapeutic potential of RD for skin fibrosis in the future.


Subject(s)
Adenosine Monophosphate , Alanine , Fibroblasts , Fibrosis , Signal Transduction , Skin , Transforming Growth Factor beta1 , Animals , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Fibrosis/drug therapy , Alanine/analogs & derivatives , Alanine/pharmacology , Alanine/therapeutic use , Fibroblasts/drug effects , Fibroblasts/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/metabolism , Mice , Skin/drug effects , Skin/pathology , Skin/metabolism , Humans , Autophagy/drug effects , Keloid/drug therapy , Keloid/metabolism , Keloid/pathology , Antiviral Agents/pharmacology , TOR Serine-Threonine Kinases/metabolism , Bleomycin , Phosphatidylinositol 3-Kinases/metabolism , Male , Proto-Oncogene Proteins c-akt/metabolism , Smad Proteins/metabolism
4.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(3): 286-295, 2024 Jun 01.
Article in English, Chinese | MEDLINE | ID: mdl-39049647

ABSTRACT

OBJECTIVES: This study aimed to investigate the temporal and spatial changes in the expression of periostin during periodontal inflammation in mice. METHODS: A periodontitis model was constructed using silk thread ligation. Mice were randomly divided into five groups including control group, 4-day ligation group, 7-day ligation group, 14-day ligation group, and self-healing group (thread removal for 14 days after 14-day ligation). Micro-CT and histological staining were performed to characterize the dynamic changes in the mouse periodontal tissue in each group. RNAscope and immunohistochemical staining were used to analyze the pattern of changes in periostin at various stages of periodontitis. The cell experiment was divided into three groups: control group, lipopolysaccharide (LPS) stimulation group (treated with LPS for 12 h), and LPS stimulation removal group (treated with LPS for 3 h followed by incubation with medium for 9 h). Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression of periostin, transforming growth factor-ß1 (TGF-ß1), and matrix metalloproteinase 2 (MMP2). RESULTS: Significant alveolar bone resorption was observed 7 days after ligation. With increasing duration of ligation, the damage to the mouse periodontal tissue was aggravated, which manifested as increased osteoclasts, widening of the periodontal membrane space, and decreased alveolar bone height. Some degree of periodontal tissue repair was observed in the self-healing group. Periostin expression decreased at 4 and 7 days compared with the control group and increased at 14 days compared with 4 and 7 days. A significant recovery was found in the self-healing group. The qRT-PCR results showed that the expression of periostin and TGF-ß1 in the LPS stimulation group decreased compared with that in the control group but significantly recovered in the LPS removal group. CONCLUSIONS: Periostin expression in the PDL of mice showed a downward and upward trend with inflammation progression. The significant recovery of periostin expression after removing inflammatory stimuli may be related to TGF-ß1, which is crucial to maintain the integrity of the PDL.


Subject(s)
Alveolar Bone Loss , Cell Adhesion Molecules , Disease Models, Animal , Lipopolysaccharides , Periodontitis , Transforming Growth Factor beta1 , Animals , Cell Adhesion Molecules/metabolism , Mice , Periodontitis/metabolism , Transforming Growth Factor beta1/metabolism , Alveolar Bone Loss/metabolism , Matrix Metalloproteinase 2/metabolism , X-Ray Microtomography , Periostin
5.
PLoS One ; 19(7): e0306775, 2024.
Article in English | MEDLINE | ID: mdl-38985836

ABSTRACT

BACKGROUND: This study evaluated the effect of microvesicles(MVs) from quiescent and TGF-ß1 stimulated hepatic stellate cells (HSC-MVs, TGF-ß1HSC-MVs) on H2O2-induced human umbilical vein endothelial cells (HUVECs) injury and CCl4-induced rat hepatic vascular injury. METHODS: HUVECs were exposed to hydrogen peroxide (H2O2) to establish a model for vascular endothelial cell injury. HSC-MVs or TGF-ß1HSC-MVs were co-cultured with H2O2-treated HUVECs, respectively. Indicators including cell survival rate, apoptosis rate, oxidative stress, migration, invasion, and angiogenesis were measured. Simultaneously, the expression of proteins such as PI3K, AKT, MEK1+MEK2, ERK1+ERK2, VEGF, eNOS, and CXCR4 was assessed, along with activated caspase-3. SD rats were intraperitoneally injected with CCl4 twice a week for 10 weeks to induce liver injury models. HSC-MVs or TGF-ß1HSC-MVs were injected into the tail vein of rats. Liver and hepatic vascular damage were also detected. RESULTS: In H2O2-treated HUVECs, HSC-MVs increased cell viability, reduced cytotoxicity and apoptosis, improved oxidative stress, migration, and angiogenesis, and upregulated protein expression of PI3K, AKT, MEK1/2, ERK1/2, VEGF, eNOS, and CXCR4. Conversely, TGF-ß1HSC-MVs exhibited opposite effects. CCl4- induced rat hepatic injury model, HSC-MVs reduced the release of ALT and AST, hepatic inflammation, fatty deformation, and liver fibrosis. HSC-MVs also downregulated the protein expression of CD31 and CD34. Conversely, TGF-ß1HSC-MVs demonstrated opposite effects. CONCLUSION: HSC-MVs demonstrated a protective effect on H2O2-treated HUVECs and CCl4-induced rat hepatic injury, while TGF-ß1HSC-MVs had an aggravating effect. The effects of MVs involve PI3K/AKT/VEGF, CXCR4, and MEK/ERK/eNOS pathways.


Subject(s)
Hepatic Stellate Cells , Human Umbilical Vein Endothelial Cells , Hydrogen Peroxide , Transforming Growth Factor beta1 , Animals , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Humans , Transforming Growth Factor beta1/metabolism , Hydrogen Peroxide/pharmacology , Rats , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Oxidative Stress/drug effects , Male , Liver/pathology , Liver/metabolism , Liver/drug effects , Liver/injuries , Rats, Sprague-Dawley , Apoptosis/drug effects , Cell-Derived Microparticles/metabolism , Cell Survival/drug effects , Carbon Tetrachloride/toxicity , Cell Movement/drug effects , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism
6.
Chem Pharm Bull (Tokyo) ; 72(7): 669-675, 2024.
Article in English | MEDLINE | ID: mdl-39010213

ABSTRACT

Tendon injury is a prevalent orthopedic disease that currently lacks effective treatment. Galangin (GLN) is a vital flavonoid found abundantly in galangal and is known for its natural activity. This study aimed to investigate the GLN-mediated molecular mechanism of tendon-derived stem cells (TDSCs) in tendon repair. The TDSCs were characterized using alkaline phosphatase staining, alizarin red S staining, oil red O staining, and flow cytometry. The effect of GLN treatment on collagen deposition was evaluated using Sirius red staining and quantitative (q)PCR, while a Western bot was used to assess protein levels and analyze pathways. Results showed that GLN treatment not only increased the collagen deposition but also elevated the mRNA expression and protein levels of multiple tendon markers like collagen type I alpha 1 (COL1A1), decorin (DCN) and tenomodulin (TNMD) in TDSCs. Moreover, GLN was also found to upregulate the protein levels of transforming growth factor ß1 (TGF-ß1) and p-Smad3 to activate the TGF-ß1/Smad3 signaling pathway, while GLN mediated collagen deposition in TDSCs was reversed by LY3200882, a TGF-ß receptor inhibitor. The study concluded that GLN-mediated TDSCs enhanced tendon repair by activating the TGF-ß1/Smad3 signaling pathway, suggesting a novel therapeutic option in treating tendon repair.


Subject(s)
Flavonoids , Signal Transduction , Smad3 Protein , Stem Cells , Tendons , Transforming Growth Factor beta1 , Flavonoids/pharmacology , Flavonoids/chemistry , Transforming Growth Factor beta1/metabolism , Signal Transduction/drug effects , Animals , Smad3 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Tendons/cytology , Tendons/metabolism , Tendons/drug effects , Rats , Cells, Cultured , Rats, Sprague-Dawley , Tendon Injuries/drug therapy , Tendon Injuries/metabolism
7.
Nat Commun ; 15(1): 5680, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971819

ABSTRACT

Obesity shapes anti-tumor immunity through lipid metabolism; however, the mechanisms underlying how colorectal cancer (CRC) cells utilize lipids to suppress anti-tumor immunity remain unclear. Here, we show that tumor cell-intrinsic ATP6V0A1 drives exogenous cholesterol-induced immunosuppression in CRC. ATP6V0A1 facilitates cholesterol absorption in CRC cells through RAB guanine nucleotide exchange factor 1 (RABGEF1)-dependent endosome maturation, leading to cholesterol accumulation within the endoplasmic reticulum and elevated production of 24-hydroxycholesterol (24-OHC). ATP6V0A1-induced 24-OHC upregulates TGF-ß1 by activating the liver X receptor (LXR) signaling. Subsequently, the release of TGF-ß1 into the tumor microenvironment by CRC cells activates the SMAD3 pathway in memory CD8+ T cells, ultimately suppressing their anti-tumor activities. Moreover, we identify daclatasvir, a clinically used anti-hepatitis C virus (HCV) drug, as an ATP6V0A1 inhibitor that can effectively enhance the memory CD8+ T cell activity and suppress tumor growth in CRC. These findings shed light on the potential for ATP6V0A1-targeted immunotherapy in CRC.


Subject(s)
CD8-Positive T-Lymphocytes , Cholesterol , Colorectal Neoplasms , Signal Transduction , Transforming Growth Factor beta1 , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Animals , Cholesterol/metabolism , Mice , Cell Line, Tumor , Transforming Growth Factor beta1/metabolism , Immunologic Memory , Vacuolar Proton-Translocating ATPases/metabolism , Tumor Microenvironment/immunology , Liver X Receptors/metabolism , Hydroxycholesterols/metabolism , Hydroxycholesterols/pharmacology , Pyrrolidines/pharmacology , Smad3 Protein/metabolism , Mice, Inbred C57BL , Carbamates/pharmacology
8.
Otol Neurotol ; 45(7): 810-817, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38995724

ABSTRACT

HYPOTHESIS: Transforming growth factor beta-1 (TGFß-1) and connective tissue growth factor (CTGF) are upregulated in the implanted human cochlea. BACKGROUND: Cochlear implantation can lead to insertion trauma and intracochlear new tissue formation, which can detrimentally affect implant performance. TGFß-1 and CTGF are profibrotic proteins implicated in various pathologic conditions, but little is known about their role in the cochlea. The present study aimed to characterize the expression of these proteins in the human implanted cochlea. METHODS: Archival human temporal bones (HTB) acquired from 12 patients with previous CI and histopathological evidence of new tissue formation as well as surgical samples of human intracochlear scar tissue surrounding the explanted CI were used in this study. Histopathologic analysis of fibrosis and osteoneogenesis was conducted using H&E. Protein expression was characterized using immunofluorescence. RNA expression from surgical specimens of fibrotic tissue surrounding the CI was quantified using qRT-PCR. RESULTS: TGFß-1 and CTGF protein expressions were upregulated in the areas of fibrosis and osteoneogenesis surrounding the CI HTB. Similarly, surgical samples demonstrated upregulation of protein and mRNA expression of TGFß-1 and mild upregulation of CTGF compared with control. TGFß-1 was expressed diffusely within the fibrous capsule, whereas CTGF was expressed in the thickened portion toward the modiolus and the fibrosis-osteoneogensis junction. CONCLUSION: To our knowledge, this is the first study to demonstrate increased expression of TGFß-1 and CTGF in the human implanted cochlea and may provide better understanding of the mechanism behind this pathogenic process to better develop future mitigating interventions.


Subject(s)
Cochlea , Connective Tissue Growth Factor , Transforming Growth Factor beta1 , Humans , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cochlea/metabolism , Male , Middle Aged , Female , Cochlear Implantation , Cochlear Implants , Temporal Bone/metabolism , Temporal Bone/pathology , Fibrosis , Aged , Adult
9.
Bull Exp Biol Med ; 177(1): 115-123, 2024 May.
Article in English | MEDLINE | ID: mdl-38963596

ABSTRACT

The cardiac perivascular niche is a cellular microenvironment of a blood vessel. The principles of niche regulation are still poorly understood. We studied the effect of TGFß1 on cells forming the cardiac perivascular niche using 3D cell culture (cardiospheres). Cardiospheres contained progenitor (c-Kit), endothelial (CD31), and mural (αSMA) cells, basement membrane proteins (laminin) and extracellular matrix proteins (collagen I, fibronectin). TGFß1 treatment decreased the length of CD31+ microvasculature, VE cadherin protein level, and proportion of NG2+ cells, and increased proportion of αSMA+ cells and transgelin/SM22α protein level. We supposed that this effect is related to the stabilizing function of TGFß1 on vascular cells: decreased endothelial cell proliferation, as shown for HUVEC, and activation of mural cell differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Cell Differentiation/drug effects , Humans , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Animals , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Cadherins/metabolism , Laminin/metabolism , Laminin/pharmacology , Muscle Proteins/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/cytology , Fibronectins/metabolism , Fibronectins/pharmacology , Antigens, CD/metabolism , Myocardium/metabolism , Myocardium/cytology , Stem Cell Niche/drug effects , Stem Cell Niche/physiology , Collagen Type I/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/cytology , Cell Culture Techniques, Three Dimensional/methods
10.
Sci Rep ; 14(1): 15998, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987293

ABSTRACT

Pathological fibrosis is a significant complication of surgical procedures resulting from the accumulation of excess collagen at the site of repair which can compromise the tissue architecture and severely impede the function of the affected tissue. Few prophylactic treatments exist to counteract this process; however, the use of amniotic membrane allografts has demonstrated promising clinical outcomes. This study aimed to identify the underlying mechanism of action by utilizing relevant models that accurately represent the pathophysiology of the disease state. This study employed a pro-fibrotic in vitro system using TGFß1 stimulation and macromolecular crowding techniques to evaluate the mechanism by which amniotic membrane allografts regulate collagen biosynthesis and deposition. Following treatment with dehydrated human amnion chorion membrane (DHACM), subsequent RNA sequencing and functional enrichment with Reactome pathway analysis indicated that amniotic membranes are indeed capable of regulating genes associated with the composition and function of the extracellular matrix. Furthermore, macromolecular crowding was used in vitro to expand the evaluation to include both the effects of DHACM and a lyophilized human amnion/chorion membrane (LHACM). DHACM and LHACM regulate the TGFß pathway and myofibroblast differentiation. Additionally, both DHACM and LHACM modulate the production, secretion, and deposition of collagen type I, a primary target for pathological fibrosis. These observations support the hypothesis that amniotic membranes may interrupt pathological fibrosis by regulating collagen biosynthesis and associated pathways.


Subject(s)
Amnion , Chorion , Collagen , Amnion/metabolism , Humans , Chorion/metabolism , Collagen/metabolism , Transforming Growth Factor beta1/metabolism , Cell Differentiation , Extracellular Matrix/metabolism , Myofibroblasts/metabolism , Fibrosis , Female , Collagen Type I/metabolism , Collagen Type I/genetics
11.
Sci Rep ; 14(1): 15947, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987362

ABSTRACT

The clinical impact of soluble molecules in pleural effusion (PE) is unclear in patients with malignant pleural mesothelioma (MPM). In this single-center, retrospective, observational study, we assessed soluble forms of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and PD-1 ligand 1 (PD-L1) using enzyme-linked immunosorbent assays; three TGF-ß isoforms were measured via multiplex assay in PE of patients with fibrinous pleuritis (FP) or MPM, to assess relationships between the levels of six molecules, clinicopathological characteristics, and efficacy of immune checkpoint inhibitors. Soluble forms of CTLA-4, PD-L1, PD-1, TGF-ß1, TGF-ß2, and TGF-ß3 were variably produced in PE of FP (n = 34) and MPM (n = 79); we found significant relationships between the six molecules and clinicopathological features. Although none of the three soluble immune checkpoint molecules showed diagnostic or prognostic effects in patients with MPM, TGF-ß2 level in PE is a useful differential diagnostic marker between FP and MPM. Both TGF-ß1 and TGF-ß3 levels are promising prognostic markers for MPM. Moreover, we found that higher baseline levels of PD-1 soluble forms predicted the response to anti-PD1 monotherapy. Our findings identify novel diagnostic, prognostic, and predictive biomarkers for anti-PD1 therapy in patients with MPM.


Subject(s)
Immune Checkpoint Proteins , Mesothelioma, Malignant , Pleural Effusion, Malignant , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , Humans , Male , Female , Mesothelioma, Malignant/metabolism , Mesothelioma, Malignant/pathology , Mesothelioma, Malignant/drug therapy , Aged , Middle Aged , Pleural Effusion, Malignant/metabolism , Pleural Effusion, Malignant/pathology , Pleural Effusion, Malignant/immunology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism , Retrospective Studies , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Transforming Growth Factor beta3/metabolism , Biomarkers, Tumor/metabolism , CTLA-4 Antigen/metabolism , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen/metabolism , Prognosis , Pleural Neoplasms/metabolism , Pleural Neoplasms/pathology , Aged, 80 and over , Programmed Cell Death 1 Receptor/metabolism , Adult
12.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38990714

ABSTRACT

Dermal fibroblasts deposit type I collagen, the dominant extracellular matrix molecule found in skin, during early postnatal development. Coincident with this biosynthetic program, fibroblasts proteolytically remodel pericellular collagen fibrils by mobilizing the membrane-anchored matrix metalloproteinase, Mmp14. Unexpectedly, dermal fibroblasts in Mmp14-/- mice commit to a large-scale apoptotic program that leaves skin tissues replete with dying cells. A requirement for Mmp14 in dermal fibroblast survival is recapitulated in vitro when cells are embedded within, but not cultured atop, three-dimensional hydrogels of crosslinked type I collagen. In the absence of Mmp14-dependent pericellular proteolysis, dermal fibroblasts fail to trigger ß1 integrin activation and instead actuate a TGF-ß1/phospho-JNK stress response that leads to apoptotic cell death in vitro as well as in vivo. Taken together, these studies identify Mmp14 as a requisite cell survival factor that maintains dermal fibroblast viability in postnatal dermal tissues.


Subject(s)
Apoptosis , Cell Survival , Fibroblasts , Matrix Metalloproteinase 14 , Animals , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/genetics , Fibroblasts/metabolism , Mice , Mice, Knockout , Collagen Type I/metabolism , Collagen Type I/genetics , Integrin beta1/metabolism , Integrin beta1/genetics , Transforming Growth Factor beta1/metabolism , Dermis/metabolism , Dermis/cytology , Cells, Cultured , Extracellular Matrix/metabolism , Mice, Inbred C57BL , Skin/metabolism
13.
Physiol Rep ; 12(13): e16148, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38991987

ABSTRACT

Pulmonary fibrosis is characterized by pathological accumulation of scar tissue in the lung parenchyma. Many of the processes that are implicated in fibrosis, including increased extracellular matrix synthesis, also occur following pneumonectomy (PNX), but PNX instead results in regenerative compensatory growth of the lung. As fibroblasts are the major cell type responsible for extracellular matrix production, we hypothesized that comparing fibroblast responses to PNX and bleomycin (BLM) would unveil key differences in the role they play during regenerative versus fibrotic lung responses. RNA-sequencing was performed on flow-sorted fibroblasts freshly isolated from mouse lungs 14 days after BLM, PNX, or sham controls. RNA-sequencing analysis revealed highly similar biological processes to be involved in fibroblast responses to both BLM and PNX, including TGF-ß1 and TNF-α. Interestingly, we observed smaller changes in gene expression after PNX than BLM at Day 14, suggesting that the fibroblast response to PNX may be muted by expression of transcripts that moderate pro-fibrotic pathways. Itpkc, encoding inositol triphosphate kinase C, was a gene uniquely up-regulated by PNX and not BLM. ITPKC overexpression in lung fibroblasts antagonized the pro-fibrotic effect of TGF-ß1. RNA-sequencing analysis has identified considerable overlap in transcriptional changes between fibroblasts following PNX and those overexpressing ITPKC.


Subject(s)
Bleomycin , Fibroblasts , Mice, Inbred C57BL , Pneumonectomy , Pulmonary Fibrosis , Bleomycin/pharmacology , Animals , Fibroblasts/metabolism , Fibroblasts/drug effects , Mice , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/cytology , Lung/pathology , Male , Sequence Analysis, RNA/methods , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cells, Cultured
14.
Mol Med ; 30(1): 99, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982366

ABSTRACT

BACKGROUND: Enhanced glycolysis is a crucial metabolic event that drives the development of liver fibrosis, but the molecular mechanisms have not been fully understood. Lactate is the endproduct of glycolysis, which has recently been identified as a bioactive metabolite binding to G-protein-coupled receptor 81 (GPR81). We then questioned whether GPR81 is implicated in the development of liver fibrosis. METHODS: The level of GPR81 was determined in mice with carbon tetrachloride (CCl4)-induced liver fibrosis and in transforming growth factor beta 1 (TGF-ß1)-activated hepatic stellate cells (HSCs) LX-2. To investigate the significance of GPR81 in liver fibrosis, wild-type (WT) and GPR81 knockout (KO) mice were exposed to CCl4, and then the degree of liver fibrosis was determined. In addition, the GPR81 agonist 3,5-dihydroxybenzoic acid (DHBA) was supplemented in CCl4-challenged mice and TGF-ß1-activated LX-2 cells to further investigate the pathological roles of GPR81 on HSCs activation. RESULTS: CCl4 exposure or TGF-ß1 stimulation significantly upregulated the expression of GPR81, while deletion of GPR81 alleviated CCl4-induced elevation of aminotransferase, production of pro-inflammatory cytokines, and deposition of collagen. Consistently, the production of TGF-ß1, the expression of alpha-smooth muscle actin (α-SMA) and collagen I (COL1A1), as well as the elevation of hydroxyproline were suppressed in GPR81 deficient mice. Supplementation with DHBA enhanced CCl4-induced liver fibrogenesis in WT mice but not in GPR81 KO mice. DHBA also promoted TGF-ß1-induced LX-2 activation. Mechanistically, GPR81 suppressed cAMP/CREB and then inhibited the expression of Smad7, a negative regulator of Smad3, which resulted in increased phosphorylation of Smad3 and enhanced activation of HSCs. CONCLUSION: GPR81 might be a detrimental factor that promotes the development of liver fibrosis by regulating CREB/Smad7 pathway.


Subject(s)
Carbon Tetrachloride , Cyclic AMP Response Element-Binding Protein , Hepatic Stellate Cells , Liver Cirrhosis , Mice, Knockout , Receptors, G-Protein-Coupled , Signal Transduction , Smad7 Protein , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/chemically induced , Mice , Cyclic AMP Response Element-Binding Protein/metabolism , Hepatic Stellate Cells/metabolism , Smad7 Protein/metabolism , Smad7 Protein/genetics , Transforming Growth Factor beta1/metabolism , Male , Humans , Cell Line , Disease Models, Animal , Mice, Inbred C57BL , Gene Deletion
15.
Bull Exp Biol Med ; 177(1): 74-78, 2024 May.
Article in English | MEDLINE | ID: mdl-38955854

ABSTRACT

Activated hepatic stellate cells differentiate into myofibroblasts, which synthesize and secrete extracellular matrix (ECM) leading to liver fibrosis. It was previously demonstrated that bulleyaconitine A (BLA), an alkaloid from Aconitum bulleyanum, inhibits proliferation and promotes apoptosis of human hepatic Lieming Xu-2 (LX-2) cells. In this study, we analyzed the effect of BLA on the production of ECM and related proteins by LX-2 cells activated with acetaldehyde (AA). The cells were randomized into the control group, AA group (cells activated with 400 µM AA), and BLA+AA group (cells cultured in the presence of 400 µM AA and 18.75 µg/ml BLA). In the BLA+AA group, the contents of collagens I and III and the expression of α-smooth muscle actin and transforming growth factor-ß1 (TGF-ß1) were statistically significantly higher than in the control, but lower than in the AA group. Expression of MMP-1 in the BLA+AA group was also significantly higher than in the AA group, but lower than in the control. Expression of TIMP-1 in the BLA+AA group was significantly higher than in the control, but lower than in the AA group. Thus, BLA suppressed activation and proliferation of LX-2 cells by inhibiting TGF-ß1 signaling pathway and decreasing the content of collagens I and III by reducing the MMP-1/TIMP-1 ratio.


Subject(s)
Acetaldehyde , Aconitine , Actins , Collagen Type I , Extracellular Matrix , Hepatic Stellate Cells , Tissue Inhibitor of Metalloproteinase-1 , Transforming Growth Factor beta1 , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Acetaldehyde/pharmacology , Acetaldehyde/analogs & derivatives , Aconitine/pharmacology , Aconitine/analogs & derivatives , Collagen Type I/metabolism , Collagen Type I/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Actins/metabolism , Actins/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Cell Line , Collagen Type III/metabolism , Collagen Type III/genetics , Cell Proliferation/drug effects , Aconitum/chemistry , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology
16.
J Agric Food Chem ; 72(28): 15715-15724, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38961631

ABSTRACT

Neohesperidin dihydrochalcone (NHDC) is a citrus-originated, seminatural sweetener. There is no investigation concerning the effect of NHDC on ulcerative colitis. The purpose of this study was to determine the therapeutic and protective effects of NHDC in Wistar Albino rats. NHDC was given for 7 days after or before colitis induction. The results showed that NHDC significantly reduced the interleukin-6 (IL-6), interleukin-10 (IL-10), transforming growth factor-ß1 (TGF-ß1), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) levels. Catalase levels did not show a significant difference between the groups. NHDC provided a remarkable decrease in the expression levels of cyclooxygenase-2 (COX-2), myeloperoxidase (MPO), malondialdehyde (MDA), 8-hydroxy-2'-deoxyguanosine (8-OHdG), and nuclear factor kappa B (NF-κB). Total antioxidant status (TAS) levels were significantly elevated in NHDC treatment groups, while total oxidant status (TOS) and oxidative stress index (OSI) levels were significantly decreased. NHDC provided remarkable improvement in histological symptoms such as epithelial erosion, edema, mucosal necrosis, inflammatory cell infiltration, and hemorrhage. Also, caspase-3 expression levels were statistically decreased in NHDC treatment groups. The results indicated that NHDC might be a protection or alternative treatment for ulcerative colitis.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Apoptosis , Chalcones , Hesperidin , NF-kappa B , Rats, Wistar , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/administration & dosage , Rats , Antioxidants/pharmacology , Male , Apoptosis/drug effects , Chalcones/pharmacology , Chalcones/administration & dosage , Hesperidin/analogs & derivatives , Hesperidin/pharmacology , Hesperidin/administration & dosage , NF-kappa B/genetics , NF-kappa B/metabolism , Humans , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Oxidative Stress/drug effects , Interleukin-6/genetics , Interleukin-6/metabolism , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/chemically induced , Malondialdehyde/metabolism , Peroxidase/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/immunology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics
17.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000533

ABSTRACT

Vascular calcification (VC) is a cardiovascular disease characterized by calcium salt deposition in vascular smooth muscle cells (VSMCs). Standard in vitro models used in VC investigations are based on VSMC monocultures under static conditions. Although these platforms are easy to use, the absence of interactions between different cell types and dynamic conditions makes these models insufficient to study key aspects of vascular pathophysiology. The present study aimed to develop a dynamic endothelial cell-VSMC co-culture that better mimics the in vivo vascular microenvironment. A double-flow bioreactor supported cellular interactions and reproduced the blood flow dynamic. VSMC calcification was stimulated with a DMEM high glucose calcification medium supplemented with 1.9 mM NaH2PO4/Na2HPO4 (1:1) for 7 days. Calcification, cell viability, inflammatory mediators, and molecular markers (SIRT-1, TGFß1) related to VSMC differentiation were evaluated. Our dynamic model was able to reproduce VSMC calcification and inflammation and evidenced differences in the modulation of effectors involved in the VSMC calcified phenotype compared with standard monocultures, highlighting the importance of the microenvironment in controlling cell behavior. Hence, our platform represents an advanced system to investigate the pathophysiologic mechanisms underlying VC, providing information not available with the standard cell monoculture.


Subject(s)
Cell Differentiation , Coculture Techniques , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Vascular Calcification , Humans , Vascular Calcification/metabolism , Vascular Calcification/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Cells, Cultured , Cell Survival , Transforming Growth Factor beta1/metabolism , Sirtuin 1/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Bioreactors
18.
Lasers Med Sci ; 39(1): 176, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38976032

ABSTRACT

Laser therapy has shown effectiveness in promoting wound healing by influencing various physiological factors such as blood flow, cytokines, histamine, nerve signals, lymphocyte function, tissue oxygenation, and cell growth. This study aims to evaluate the therapeutic efficacy of Photobiomodulation (PBM) treatment, by using diode laser, in modifying the levels of interleukin-1 beta (IL1ß) and transforming growth factor beta-1 (TGFß-1) in patients diagnosed with aphthous stomatitis. A before-after interventional design was conducted over 10 months with 20 subjects. Data on demographic details and serum concentrations of IL1ß and TGFß-1 were collected pre-treatment and on Days 3 and 7 post-treatments. The intervention involved a single session of four 30-second applications of a QuickLase dual-wavelength laser operating at 980 nm. Results show significant reductions in IL1ß and TGFß-1 levels after 7 days of treatment, indicating a time-dependent effect of PBM therapy on these inflammatory markers. The findings suggest that PBM therapy holds promise as an intervention for reducing inflammation associated with aphthous stomatitis.


Subject(s)
Interleukin-1beta , Lasers, Semiconductor , Low-Level Light Therapy , Stomatitis, Aphthous , Transforming Growth Factor beta1 , Humans , Interleukin-1beta/blood , Low-Level Light Therapy/methods , Adult , Female , Male , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/metabolism , Stomatitis, Aphthous/radiotherapy , Stomatitis, Aphthous/therapy , Lasers, Semiconductor/therapeutic use , Middle Aged , Young Adult
19.
FASEB J ; 38(13): e23756, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38949649

ABSTRACT

Asthma is a chronic pulmonary disease with the worldwide prevalence. The structural alterations of airway walls, termed as "airway remodeling", are documented as the core contributor to the airway dysfunction during chronic asthma. Forkhead box transcription factor FOXK2 is a critical regulator of glycolysis, a metabolic reprogramming pathway linked to pulmonary fibrosis. However, the role of FOXK2 in asthma waits further explored. In this study, the chronic asthmatic mice were induced via ovalbumin (OVA) sensitization and repetitive OVA challenge. FOXK2 was upregulated in the lungs of OVA mice and downregulated after adenovirus-mediated FOXK2 silencing. The lung inflammation, peribronchial collagen deposition, and glycolysis in OVA mice were obviously attenuated after FOXK2 knockdown. Besides, the expressions of FOXK2 and SIRT2 in human bronchial epithelial cells (BEAS-2B) were increasingly upregulated upon TGF-ß1 stimulation and downregulated after FOXK2 knockdown. Moreover, the functional loss of FOXK2 remarkably suppressed TGF-ß1-induced epithelial-mesenchymal transition (EMT) and glycolysis in BEAS-2B cells, as manifested by the altered expressions of EMT markers and glycolysis enzymes. The glycolysis inhibitor 2-deoxy-d-glucose (2-DG) inhibited the EMT in TGF-ß1-induced cells, making glycolysis a driver of EMT. The binding of FOXK2 to SIRT2 was validated, and SIRT2 overexpression blocked the FOXK2 knockdown-mediated inhibition of EMT and glycolysis in TGF-ß1-treated cells, which suggests that FOXK2 regulates EMT and glycolysis in TGF-ß1-treated cells in a SIRT2-dependnet manner. Collectively, this study highlights the protective effect of FOXK2 knockdown on airway remodeling during chronic asthma.


Subject(s)
Airway Remodeling , Asthma , Forkhead Transcription Factors , Glycolysis , Sirtuin 2 , Asthma/metabolism , Asthma/pathology , Animals , Sirtuin 2/metabolism , Sirtuin 2/genetics , Mice , Airway Remodeling/physiology , Humans , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Epithelial-Mesenchymal Transition , Mice, Inbred BALB C , Female , Transforming Growth Factor beta1/metabolism , Lung/metabolism , Lung/pathology , Cell Line
20.
Sci Rep ; 14(1): 16966, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043870

ABSTRACT

Stem cells' differentiation toward cardiac lineage is a complex process dependent on various alterations in molecular basis and regulation pathways. The aim of the study is to show that endometrium-derived stromal cells - menstrual, endometrial and endometriotic, could be an attractive source for examination of the mechanisms underlying cardiomyogenesis. After treatment with Decitabine, Angiotensin II and TGF-ß1, cells demonstrated morphological dedifferentiation into early cardiomyocyte-like cells and expressed CD36, CD106, CD172a typically used to sort for human pluripotent stem cell-derived cardiomyocytes. RT-qPCR revealed changed cells' genetic profiles, as majority of cardiac lineage differentiation related genes and cardiac ion channels (calcium, sodium, potassium) coding genes were upregulated after 6 and 13 days of exposure. Additionally, analysis of expression of various signaling proteins (FOXO1, PDGFB, TGFBR1, mTOR, VEGFA, WNT4, Notch1) coding genes showed differences between cell cultures as they seem to employ distinct signaling pathways through differentiation initiation. Early stages of differentiation had biggest impact on cardiomyogenesis related proteins (Nkx-2.5, EZH2, FOXO3a, H3K9Ac) levels, as we noticed after conducting Western blot and as expected, early cardiac transcription factor Nkx-2.5 was highly expressed and localized in nucleus of differentiating cells. These findings led us to assess endometrium origin stromal cells' potential to differentiate towards cardiomyogenic lineage and better understand the regulation of complex differentiation processes in ex vivo model systems.


Subject(s)
Angiotensin II , Cell Differentiation , Decitabine , Endometrium , Myocytes, Cardiac , Stromal Cells , Transforming Growth Factor beta1 , Humans , Female , Cell Differentiation/drug effects , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Endometrium/cytology , Endometrium/metabolism , Endometrium/drug effects , Stromal Cells/metabolism , Stromal Cells/drug effects , Stromal Cells/cytology , Angiotensin II/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Decitabine/pharmacology , Cells, Cultured , Adult , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL