Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52.054
Filter
1.
Mol Biol Rep ; 51(1): 1031, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352573

ABSTRACT

BACKGROUND: Fatty liver disease is a metabolic disorder that recently has been classified into two categories: metabolic dysfunction-associated fatty liver disease (MAFLD) and non-MAFLD. TGF-ß signaling pathway is likely a significant factor in the pathogenesis of this condition, exerting its effects through its downstream signaling proteins, Smad2/3. Accordingly, this study aimed to investigate the TGF-ß signaling pathway in the white blood cells (WBCs) of patients with MAFLD compared to those with non-MAFLD and control groups. METHODS AND RESULTS: In this study, 41 patients with fatty liver were evaluated, comprising 22 patients with MAFLD and 19 patients with non-MAFLD, and compared to 22 healthy controls. Gene expression of TGF-ß1, TGF-ß3, and CTGF were quantified using qRT-PCR, and the protein expressions of Smad2/3 and P-Smad2/3 were analyzed using western blotting. Gene expression analysis revealed a significant decrease in the gene expressions of the TGF-ß1 and TGF-ß3 and an increase in CTGF gene expression in patients with MAFLD and non-MAFLD compared to the control group. Notably, the Smad2/3 protein expression was significantly higher in the non-MAFLD group compared to the control group (P < 0.05). On the other hand, the P-smad2/3 protein expression was significantly elevated in the MAFLD group compared to the control group (P < 0.001). CONCLUSIONS: TGF-ß signaling pathway in WBCs of patients with fatty liver are affected by a complex signaling pathway. However, metabolic factors most probably affect TGF-ß1 gene expression and its downstream signaling proteins more than TGF-ß3.


Subject(s)
Connective Tissue Growth Factor , Signal Transduction , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta1 , Humans , Male , Case-Control Studies , Female , Middle Aged , Adult , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Fatty Liver/metabolism , Fatty Liver/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Smad Proteins/metabolism , Smad Proteins/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Gene Expression Regulation
2.
Cell Mol Biol (Noisy-le-grand) ; 70(9): 136-141, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39380267

ABSTRACT

This study aimed to explore the influence and mechanism of low-intensity pulsed ultrasound (LIPUS) combined with Rhodiola bone penetration on the formation of spinal fusion bone. Sixty clean-grade New Zealand white rabbits were selected for randomization and divided into combined group and Rhodiola group, with 30 rabbits in each group to construct a rabbit lumbar intervertebral fusion model, using Rhodiola intervention and Rhodiola combined with LIPUS intervention protocol, respectively. The axial strength, axial stiffness, maximum compressive load, vascular endothelial growth factor (VEGF), cycloxygenase-2 (COX-2), prostaglandin E2 (PGE2) and transforming growth factor-ß (TGF-ß) were compared after HE staining, immunohistochemistry and biomechanical detection. Spine fusion rate was 100.00%; the combined bone graft tissue had implanted bone cell degeneration, cell necrosis and cell hyperplasia, chondrocytes differentiated into trabecular bone and some hematopoietic cells, severe cell necrosis and fiber cell proliferation and late bone formation in the Rhodiola group, VEGF, COX-2, PGE2, TGF-ß, axial strength, axial stiffness, and maximum compression load in the combined group significantly increased (P<0.05). Spinal fusion using LIPUS combined with Rhodiola can enhance biomechanical properties and promote the role of PGE2, COX-2, VEGF, TGF-ß expression and bone formation, and this protocol is worthy of clinical application.


Subject(s)
Osteogenesis , Rhodiola , Spinal Fusion , Animals , Rabbits , Rhodiola/chemistry , Osteogenesis/drug effects , Spinal Fusion/methods , Dinoprostone/metabolism , Ultrasonic Waves , Vascular Endothelial Growth Factor A/metabolism , Cyclooxygenase 2/metabolism , Biomechanical Phenomena/drug effects , Transforming Growth Factor beta/metabolism
3.
Sci Rep ; 14(1): 23117, 2024 10 04.
Article in English | MEDLINE | ID: mdl-39367011

ABSTRACT

Psychotic and mood disorders are discussed as part of the same continuum. The potential role of immune dysregulation in defining their clinical presentations, however, remains unclear. Differences in TNF-α, IL-6 and TGF-ß levels were investigated in 143 patients with schizophrenia (SCH = 63) and bipolar disorder (BD = 80), in remission. Cytokines were evaluated against the dimensional assessment of psychosis and affective symptoms using the schizo-bipolar scale, together with the severity of the same symptom domains measured by the brief psychiatric rating scale (BPRS). Lower TGF-ß was associated with more lifetime episodes, family risk for psychosis, and more severe mood and psychotic symptoms in all patients. BPRS Affect symptoms domain correlated with lower TGF-ß levels in BD, and higher TGF-ß levels in SCH patients. Using moderated mediation analysis, TGF-ß was a relevant predictor only in the setting of non-categorical symptom distribution, with familial risk for psychosis confirmed as a significant moderator. Severity of BPRS Affect symptoms domain was an independent predictor of inclination towards the psychosis spectrum. The underlying immune dysregulation may be shared by the disorders, rather than a unique characteristic of each, having significant implications for our understanding of the continuum vs. categorical approach to psychosis and mood disorders.


Subject(s)
Bipolar Disorder , Interleukin-6 , Psychotic Disorders , Schizophrenia , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha , Humans , Female , Male , Adult , Transforming Growth Factor beta/blood , Psychotic Disorders/blood , Interleukin-6/blood , Tumor Necrosis Factor-alpha/blood , Schizophrenia/blood , Schizophrenia/immunology , Bipolar Disorder/blood , Bipolar Disorder/immunology , Middle Aged , Affect , Mood Disorders/blood , Young Adult
4.
FASEB J ; 38(19): e70077, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39370849

ABSTRACT

In idiopathic pulmonary fibrosis (IPF), epithelial abnormalities are present including bronchiolization and alveolar cell dysfunction. We hypothesized that the IPF microenvironment disrupts normal epithelial growth and differentiation. We mimicked the soluble factors within an IPF microenvironment using an IPF cocktail (IPFc), composed of nine factors which are increased in IPF lungs (CCL2, IL-1ß, IL-4, IL-8, IL-13, IL-33, TGF-ß, TNFα, and TSLP). Using IPFc, we asked whether the soluble factor milieu in IPF affects epithelial growth and differentiation and how IPFc compares to TGF-ß alone. Epithelial growth and differentiation were studied using mouse lung organoids (primary Epcam+ epithelial cells co-cultured with CCL206 fibroblasts). Organoids exposed to IPFc and TGF-ß were re-sorted into epithelial and fibroblast fractions and subjected to RNA sequencing. IPFc did not affect the number of organoids formed. However, pro-surfactant protein C expression was decreased. On these parameters, TGF-ß alone had similar effects. However, RNA sequencing of re-sorted organoids revealed that IPFc and TGF-ß had distinct effects on both epithelial cells and fibroblasts. IPFc upregulated goblet cell markers, whereas these were inhibited by TGF-ß. Although both IPFc and TGF-ß increased extracellular matrix gene expression, only TGF-ß increased myofibroblast markers. VEGF-C and Wnt signaling were among the most differentially regulated signaling pathways by IPFc versus TGF-ß. Interestingly, Wnt pathway activation rescued Sftpc downregulation induced by IPFc. In conclusion, IPFc alters epithelial differentiation in a way that is distinct from TGF-ß. Alterations in Wnt signaling contribute to these effects. IPFc may be a more comprehensive representation of the soluble factor microenvironment in IPF.


Subject(s)
Cell Differentiation , Epithelial Cells , Idiopathic Pulmonary Fibrosis , Transforming Growth Factor beta , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Animals , Mice , Epithelial Cells/metabolism , Transforming Growth Factor beta/metabolism , Humans , Organoids/metabolism , Organoids/pathology , Lung/metabolism , Lung/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Wnt Signaling Pathway , Mice, Inbred C57BL , Cells, Cultured
5.
Exp Dermatol ; 33(10): e15186, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39367569

ABSTRACT

Ubiquitin C-terminal hydrolase L1 (UCHL1) plays vital roles in cell proliferation, angiogenesis, inflammation and oxidative stress. Nevertheless, it is unclear whether UCHL1 could regulate the biologic behaviour of cells and ultimately influences wound healing. We aim to illustrate the roles and the underlying mechanism of UCHL1 in cutaneous wound healing. Murine full-thickness excisional wound model was utilised to study the effects of UCHL1 on wound healing through topical administration of the UCHL1 inhibitor LDN57444, followed by assessment of wound areas and histological alterations. Subsequently, ethynyldeoxyuridine, scratch and transwell assays were performed to examine fibroblast migration and proliferation. The extracellular matrix (ECM)-related genes expression and transforming growth factor-ß (TGF-ß)/Smad signalling pathways activation were investigated by immuno-fluorescent staining, Western blots and quantitative reverse transcription polymerase chain reaction. We identified elevated UCHL1 expression in non-healing wound tissues. The UCHL1 expression displayed a dynamic change and reached a peak on Day-7 post-wounding during the healing process in mice. Cutaneous administration of LDN57444 promoted wound healing by facilitating collagen deposition, myofibroblast activation and angiogenesis. In vitro experiments demonstrated that UCHL1 concentration dependently inhibited migration, ECM synthesis and activation of human dermal fibroblasts, which was mechanistically related to downregulation of TGF-ß/Smad signalling. Furthermore, these effects could be reversed by TGF-ß inhibitor SB431542. Our findings reveal that UCHL1 is a negative regulator of cutaneous wound healing and considered as a novel prospective therapeutic target for effective wound healing.


Subject(s)
Cell Movement , Fibroblasts , Signal Transduction , Smad Proteins , Transforming Growth Factor beta , Ubiquitin Thiolesterase , Wound Healing , Animals , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Fibroblasts/metabolism , Wound Healing/drug effects , Signal Transduction/drug effects , Mice , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Cell Movement/drug effects , Skin/metabolism , Skin/pathology , Cell Proliferation/drug effects , Dioxoles/pharmacology , Male , Humans , Benzamides/pharmacology , Extracellular Matrix/metabolism
6.
FASEB J ; 38(19): e70053, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39373847

ABSTRACT

Liver fibrosis is characterized by a wound-healing response and may progress to liver cirrhosis and even hepatocellular carcinoma. Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a tumor suppressor that participates in malignant diseases. However, the role of LHPP in liver fibrosis has not been determined. Herein, the function and regulatory network of LHPP were explored in liver fibrosis. The expression of LHPP in human and murine fibrotic liver tissues was assessed via immunohistochemistry and Western blot analysis. In addition, liver fibrosis was induced in wild-type (WT) and LHPP-/- (KO) mice after carbon tetrachloride (CCl4) or thioacetamide (TAA) treatment. The effect of LHPP was systematically assessed by using specimens acquired from the above murine models. The functional role of LHPP was further explored by detecting the pathway activity of TGF-ß/Smad3 and apoptosis after interfering with LHPP in vitro. To explore whether the function of LHPP depended on the TGF-ß/Smad3 pathway in vivo, an inhibitor of the TGF-ß/Smad3 pathway was used in CCl4-induced WT and KO mice. LHPP expression was downregulated in liver tissue samples from fibrosis patients and fibrotic mice. LHPP deficiency aggravated CCl4- and TAA-induced liver fibrosis. Moreover, through immunoblot analysis, we identified the TGF-ß/Smad3 pathway as a key downstream pathway of LHPP in vivo and in vitro. The effect of LHPP deficiency was reversed by inhibiting the TGF-ß/Smad3 pathway in liver fibrosis. These results revealed that LHPP deficiency exacerbates liver fibrosis through the TGF-ß/Smad3 pathway. LHPP may be a potential therapeutic target in hepatic fibrosis.


Subject(s)
Inorganic Pyrophosphatase , Liver Cirrhosis , Mice, Knockout , Signal Transduction , Smad3 Protein , Transforming Growth Factor beta , Animals , Smad3 Protein/metabolism , Smad3 Protein/genetics , Mice , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Humans , Inorganic Pyrophosphatase/metabolism , Inorganic Pyrophosphatase/genetics , Transforming Growth Factor beta/metabolism , Male , Mice, Inbred C57BL , Apoptosis , Carbon Tetrachloride/toxicity
7.
Front Immunol ; 15: 1430187, 2024.
Article in English | MEDLINE | ID: mdl-39351229

ABSTRACT

Increased MMP-9 expression in the tumor microenvironment (TME) plays a crucial role in the extracellular matrix remodeling to facilitate cancer invasion and metastasis. However, the mechanism of MMP-9 upregulation in TME remains elusive. Since TGF-ß and TNF-α levels are elevated in TME, we asked whether these two agents interacted to induce/augment MMP-9 expression. Using a well-established MDA-MB-231 breast cancer model, we found that the synergy between TGF-ß and TNF-α led to MMP-9 upregulation at the transcriptional and translational levels, compared to treatments with each agent alone. Our in vitro findings are corroborated by co-expression of elevated MMP-9 with TGF-ß and TNF-α in human breast cancer tissues. Mechanistically, we found that the MMP-9 upregulation driven by TGF-ß/TNF-α cooperativity was attenuated by selective inhibition of the TGF-ßRI/Smad3 pathway. Comparable outcomes were observed upon inhibition of TGF-ß-induced phosphorylation of Smad2/3 and p38. As expected, the cells defective in Smad2/3 or p38-mediated signaling did not exhibit this synergistic induction of MMP-9. Importantly, the inhibition of histone methylation but not acetylation dampened the synergistic MMP-9 expression. Histone modification profiling further identified the H3K36me2 as an epigenetic regulatory mark of this synergy. Moreover, TGF-ß/TNF-α co-stimulation led to increased levels of the transcriptionally permissive dimethylation mark at H3K36 in the MMP-9 promoter. Comparable outcomes were noted in cells deficient in NSD2 histone methyltransferase. In conclusion, our findings support a cooperativity model in which TGF-ß could amplify the TNF-α-mediated MMP-9 production via chromatin remodeling and facilitate breast cancer invasion and metastasis.


Subject(s)
Breast Neoplasms , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 9 , Neoplasm Metastasis , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha , Humans , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Tumor Necrosis Factor-alpha/metabolism , Female , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Histones/metabolism , Methylation , Signal Transduction , Tumor Microenvironment
9.
Cell Death Dis ; 15(10): 715, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39353900

ABSTRACT

The metastasis of hepatocellular carcinoma (HCC) poses a significant threat to the survival of patients. G protein-coupled receptor 56 (GPR56) has garnered extensive attention within malignant tumor research and plays a crucial role in cellular surface signal transmission. Nonetheless, its precise function in HCC remains ambiguous. Our investigation reveals a notable rise in GPR56 expression levels in human HCC cases, with heightened GPR56 levels correlating with unfavorable prognoses. GPR56 regulates TGF-ß pathway by interacting with TGFBR1, thereby promoting HCC metastasis. At the same time, GPR56 is subject to regulation by the canonical cascade of TGF-ß signaling, thereby establishing a positive feedback loop. Furthermore, the combination application of TGFBR1 inhibitor galunisertib (GAL) and GPR56 inhibitor Dihydromunduletone (DHM), significantly inhibits HCC metastasis. Interventions towards this signaling pathway could offer a promising therapeutic approach to effectively impede the metastasis of GPR56-mediated HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Neoplasm Metastasis , Receptor, Transforming Growth Factor-beta Type I , Receptors, G-Protein-Coupled , Signal Transduction , Transforming Growth Factor beta , Humans , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Transforming Growth Factor beta/metabolism , Animals , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Cell Line, Tumor , Mice , Mice, Nude , Quinolines/pharmacology , Gene Expression Regulation, Neoplastic , Male , Pyrazoles
10.
Lasers Med Sci ; 39(1): 230, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39222167

ABSTRACT

The aim of the present study was to evaluate the photothermal effects of a subdermal high-power diode laser at a wavelength (λ) of 1470 nm in the skin of rats. Twenty male Wistar rats were used, divided into 2 groups: placebo laser (PL) and active laser (AL). A high-power diode laser equipment was applied to 5 subdermal vectors on the animal's back region. The results demonstrated that active laser animals showed a better arrangement of collagen fiber bands, an increase in the thickness of the dermis and the number of vessels. Furthermore, animals treated with active laser showed an increased immunoexpression of TGF-ß and VEGF compared to the placebo. The present work demonstrated that the subdermal high-power diode laser increases the vascularization and the expression of factors that enhance skin regeneration and may be promising resource in the esthetic and dermatology clinical treatment of skin rejuvenation.


Subject(s)
Lasers, Semiconductor , Rats, Wistar , Skin , Animals , Male , Rats , Lasers, Semiconductor/therapeutic use , Skin/radiation effects , Skin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Transforming Growth Factor beta/metabolism , Rejuvenation , Models, Animal
11.
Cell Death Dis ; 15(9): 666, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261464

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer-related mortality worldwide. Although CRC patients' survival is improved with surgical resection and immunotherapy, metastasis and recurrence remain major problems leading to poor prognosis. Therefore, exploring pathogenesis and identifying specific biomarkers are crucial for CRC early diagnosis and targeted therapy. CCDC113, a member of CCDC families, has been reported to play roles in ciliary assembly, ciliary activity, PSCI, asthma and early lung cancer diagnosis. However, the functions of CCDC113 in CRC still remain unclear. In this study, we find that CCDC113 is significantly highly expressed in CRC. High expression of CCDC113 is significantly correlated with CRC patients' poor prognosis. CCDC113 is required for CRC tumorigenesis and metastasis. RNA-seq and TCGA database analysis indicate that CCDC113 is positively correlated with TGF-ß signaling pathway. TGF-ß signaling pathway inhibitor galunisertib could reverse the increased proliferation and migration ability of CRC cells caused by CCDC113 overexpression in vitro and in vivo. These results indicate that CCDC113 promotes CRC tumorigenesis and metastasis via TGF-ß signaling pathway. In conclusion, it is the first time to explore the functions and mechanisms of CCDC113 in CRC tumorigenesis and metastasis. And CCDC113 may be a potential biomarker and therapeutic target for CRC intervention.


Subject(s)
Carcinogenesis , Cell Proliferation , Colorectal Neoplasms , Signal Transduction , Transforming Growth Factor beta , Animals , Female , Humans , Male , Mice , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Prognosis , Pyrazoles/pharmacology , Quinolines/pharmacology , Transforming Growth Factor beta/metabolism
12.
Nat Commun ; 15(1): 7996, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266508

ABSTRACT

Advancements in human-engineered heart tissue have enhanced the understanding of cardiac cellular alteration. Nevertheless, a human model simulating pathological remodeling following myocardial infarction for therapeutic development remains essential. Here we develop an engineered model of myocardial repair that replicates the phased remodeling process, including hypoxic stress, fibrosis, and electrophysiological dysfunction. Transcriptomic analysis identifies nine critical signaling pathways related to cellular fate transitions, leading to the evaluation of seventeen modulators for their therapeutic potential in a mini-repair model. A scoring system quantitatively evaluates the restoration of abnormal electrophysiology, demonstrating that the phased combination of TGFß inhibitor SB431542, Rho kinase inhibitor Y27632, and WNT activator CHIR99021 yields enhanced functional restoration compared to single factor treatments in both engineered and mouse myocardial infarction model. This engineered heart tissue repair model effectively captures the phased remodeling following myocardial infarction, providing a crucial platform for discovering therapeutic targets for ischemic heart disease.


Subject(s)
Dioxoles , Fibrosis , Myocardial Infarction , Pyridines , Tissue Engineering , Animals , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Mice , Humans , Pyridines/pharmacology , Pyridines/therapeutic use , Tissue Engineering/methods , Dioxoles/pharmacology , Dioxoles/therapeutic use , Myocardium/pathology , Myocardium/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Disease Models, Animal , Signal Transduction , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Ventricular Remodeling/drug effects , Transforming Growth Factor beta/metabolism , Heart/physiopathology , Heart/drug effects , Amides
13.
Nat Commun ; 15(1): 7941, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266532

ABSTRACT

Dedifferentiated liposarcoma (DDLPS) is the most frequent high-grade soft tissue sarcoma subtype. It is characterized by a component of undifferentiated tumor cells coexisting with a component of well-differentiated adipocytic tumor cells. Both dedifferentiated (DD) and well-differentiated (WD) components exhibit MDM2 amplification, however their cellular origin remains elusive. Using single-cell RNA sequencing, DNA sequencing, in situ multiplex immunofluorescence and functional assays in paired WD and DD components from primary DDLPS tumors, we characterize the cellular heterogeneity of DDLPS tumor and micro-environment. We identify a population of tumor adipocyte stem cells (ASC) showing striking similarities with adipocyte stromal progenitors found in white adipose tissue. We show that tumor ASC harbor the ancestral genomic alterations of WD and DD components, suggesting that both derive from these progenitors following clonal evolution. Last, we show that DD tumor cells keep important biological properties of ASC including pluripotency and that their adipogenic properties are inhibited by a TGF-ß-high immunosuppressive tumor micro-environment.


Subject(s)
Adipocytes , Clonal Evolution , Liposarcoma , Proto-Oncogene Proteins c-mdm2 , Tumor Microenvironment , Humans , Liposarcoma/genetics , Liposarcoma/pathology , Liposarcoma/metabolism , Adipocytes/pathology , Adipocytes/metabolism , Tumor Microenvironment/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Single-Cell Analysis , Female , Cell Dedifferentiation/genetics , Male , Cell Differentiation/genetics , Transforming Growth Factor beta/metabolism , Middle Aged , Aged
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1459-1466, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276041

ABSTRACT

OBJECTIVE: To investigate the mechanism by which conbercept reverses transforming growth factor-ß2 (TGF-ß2)-induced epithelial-mesenchymal transition (EMT) in human lens epithelial cells (HLECs). METHODS: Cultured HLEC SRA01/04 cells were treated with TGF-ß2, conbercept, or both, and the changes in cell proliferation, apoptosis, and migration were observed using MTT assay, flow cytometry, scratch assay, and Transwell assay. Western blotting and qRT-PCR were used to detect the changes in the expression of EMT-related epithelial cell markers (E-Cadherin, α-SMA, and Snail), extracellular matrix components, and genes related to the TGF-ß/Smad signaling pathway. RESULTS: Conbercept significantly reduced TGF-ß2-induced EMT of SRA01/04 cells, decreased the expression levels of mesenchymal and extracellular matrix markers α-SMA, Snail, collagen I, collagen IV, and FN1, and upregulated the protein and mRNA expressions of E-cadherin (P <0.05). Transwell assay showed significantly lower cell migration ability in TGF-ß2+conbercept group than in TGF-ß2 group (P <0.05). Conbercept also inhibited the increase in Smad2/3 phosphorylation levels in HLEC-SRA01/04 cells with TGF-ß2-induced EMT (P <0.01). CONCLUSION: Conbercept inhibits TGF-ß2 induced EMT by downregulating the expression of pSmad2/3 in TGF-ß/Smad signaling pathway, indicating a potential therapeutic strategy against visual loss induced by posterior capsule opacification.


Subject(s)
Cell Proliferation , Epithelial Cells , Epithelial-Mesenchymal Transition , Lens, Crystalline , Signal Transduction , Smad Proteins , Transforming Growth Factor beta2 , Humans , Epithelial-Mesenchymal Transition/drug effects , Signal Transduction/drug effects , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Transforming Growth Factor beta2/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Smad Proteins/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Cadherins/metabolism , Apoptosis/drug effects , Transforming Growth Factor beta/metabolism , Cell Line , Smad2 Protein/metabolism
15.
Allergol Immunopathol (Madr) ; 52(5): 36-43, 2024.
Article in English | MEDLINE | ID: mdl-39278849

ABSTRACT

The aim of this study was to elucidate the therapeutic effect of simvastatin on experimental autoimmune encephalomyelitis (EAE) by regulating the balance between Th17 and Treg cells in mice. C57BL/6 mice were randomly divided into four groups: normal group, EAE group, simvastatin (2 and 10 mg/kg) group, and AG490 group (with AG490 serving as the positive control). Neurological function scores of mice were assessed daily. The four groups received treatments of normal saline, normal saline, and simvastatin (2 and 10 mg/kg), respectively. In the AG490 group, mice were injected intraperitoneally with AG490 (1 mg) every other day, and treatment was halted after 3 weeks. The spinal cord was stained with hematoxylin and eosin (H&E), and immunohistochemical staining for retinoic acid receptor-related orphan receptor γ(RORγ) and Foxp3 (Foxp3) was performed. Spleen samples were taken for Th17 and Treg analysis using flow cytometry. The levels of interleukin-17 and transforming growth factor-ß (TGF-ß) were detected using enzyme-linked immunosorbent assay (ELISA). In the simvastatin and AG490 groups, recovery from neurological impairment was earlier compared to the EAE group, and the symptoms were notably improved. Both simvastatin and AG490 reduced focal inflammation, decreased RORγ-positive cell infiltration, and significantly increased the number of FOXP3-positive cells. The number of Th17 cells and the level of IL-17 in the spleen were decreased in the simvastatin and AG490 treatment groups, while the number of Treg cells and TGF-ß levels were significantly increased across all treatment groups. Simvastatin exhibits anti-inflammatory and immunomodulatory effects, potentially alleviating symptoms of neurological dysfunction of EAE. Regulating the balance between Th17 and Treg may represent a therapeutic mechanism for simvastatin in treating EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Simvastatin , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Th17 Cells/immunology , Th17 Cells/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Simvastatin/pharmacology , Simvastatin/administration & dosage , Mice , Female , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Interleukin-17/metabolism , Forkhead Transcription Factors/metabolism , Spinal Cord/immunology , Spinal Cord/drug effects , Spinal Cord/pathology , Humans , Transforming Growth Factor beta/metabolism , Disease Models, Animal
16.
Allergol Immunopathol (Madr) ; 52(5): 51-58, 2024.
Article in English | MEDLINE | ID: mdl-39278851

ABSTRACT

Allergic asthma is an important public health problem and is a complicated respiratory sickness that is characterized by bronchial inflammation, bronchoconstriction, and breathlessness. Asthma is orchestrated by type 2 immune response and remodeling is one of the important outputted problem in chronic asthma. Thymol is a naturally occurring monocyclic phenolic, it has a series of biological properties, and its immunomodulatory and anti-remodeling effects on allergic asthma were evaluated. The OVA-LPS-induced asthmatic mice were treated with thymol. Methacholine challenge test, eosinophil count, and levels of IL-4, IL-5, IL-13, and IL-33 in bronchoalveolar lavage fluid, total and OVA-specific IgE levels in serum, remodeling factors, gene expression of TGF-ß, Smad2, Smad3, and lung histopathology were done. Treatment with thymol could control AHR, eosinophil percentage levels of Th2 cytokines and Igs, remodeling factors, expression of TGF-ß, Smad2 and Smad3 genes, inflammation, goblet cell hyperplasia, and mucus production in asthmatic mice. Thymol can control asthma pathogens and related remodeling and fibrosis bio-factors and can be a potential treatment of asthma.


Subject(s)
Airway Remodeling , Asthma , Disease Models, Animal , Mice, Inbred BALB C , Signal Transduction , Smad3 Protein , Thymol , Transforming Growth Factor beta , Animals , Thymol/pharmacology , Asthma/immunology , Asthma/drug therapy , Airway Remodeling/drug effects , Airway Remodeling/immunology , Smad3 Protein/metabolism , Mice , Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Cytokines/metabolism , Female , Ovalbumin/immunology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/cytology , Eosinophils/immunology , Eosinophils/drug effects , Humans , Immunoglobulin E/immunology , Immunoglobulin E/blood , Smad2 Protein/metabolism
17.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273322

ABSTRACT

IL-15 is a homeostatic cytokine for human T and NK cells. However, whether other cytokines influence the effect of IL-15 is not known. We studied the impact that IL-10, TGF-ß, IL-17A, and IFN-γ have on the IL-15-induced proliferation of human T cells and the expression of HLA class I (HLA-I) molecules. Peripheral blood lymphocytes (PBLs) were labeled with CFSE and stimulated for 12 days with IL-15 in the absence or presence of the other cytokines. The proportion of proliferating T cells and the expression of cell surface HLA-I molecules were analyzed using flow cytometry. The IL-15-induced proliferation of T cells was paralleled by an increase in the expression of HC-10-reactive HLA-I molecules, namely on T cells that underwent ≥5-6 cycles of cell division. It is noteworthy that the IL-15-induced proliferation of T cells was potentiated by IL-10 and TGF-ß but not by IL-17 or IFN-γ and was associated with a decrease in the expression of HC-10-reactive molecules. The cytokines IL-10 and TGF-ß potentiate the proliferative capacity that IL-15 has on human T cells in vitro, an effect that is associated with a reduction in the amount of HC-10 reactive HLA class I molecules induced by IL-15.


Subject(s)
Cell Proliferation , Histocompatibility Antigens Class I , Interferon-gamma , Interleukin-10 , Interleukin-15 , Interleukin-17 , T-Lymphocytes , Transforming Growth Factor beta , Humans , Cell Proliferation/drug effects , Interferon-gamma/pharmacology , Interferon-gamma/metabolism , Interleukin-17/pharmacology , Interleukin-17/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Interleukin-10/metabolism , Interleukin-15/pharmacology , Interleukin-15/metabolism , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/cytology , Cells, Cultured , Lymphocyte Activation/drug effects
18.
Molecules ; 29(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39274908

ABSTRACT

Extended exposure to UVB (280-315 nm) radiation results in oxidative damage and inflammation of the skin. Previous research has demonstrated that pilose antler extracts have strong anti-inflammatory properties and possess antioxidant effects. This study aimed to elucidate the mechanism of pilose antler protein in repairing photodamage caused by UVB radiation in HaCaT cells and ICR mice. Pilose antler protein (PAP) was found to increase the expression of type I collagen and hyaluronic acid in HaCaT cells under UVB irradiation while also inhibiting reactive oxygen species (ROS) production and oxidative stress in vitro. In vivo, the topical application of pilose antler protein effectively attenuated UVB-induced skin damage in ICR mice by reducing interleukin-1ß (IL-ß), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) and inhibiting skin inflammation while alleviating UVB-induced oxidative stress. It was shown that pilose antler protein repaired UVB-induced photodamage through the MAPK and TGF-ß/Smad pathways.


Subject(s)
Antlers , HaCaT Cells , Mice, Inbred ICR , Oxidative Stress , Reactive Oxygen Species , Skin , Ultraviolet Rays , Ultraviolet Rays/adverse effects , Animals , Humans , Antlers/chemistry , Mice , Oxidative Stress/drug effects , Skin/drug effects , Skin/radiation effects , Skin/pathology , Skin/metabolism , Reactive Oxygen Species/metabolism , Collagen Type I/metabolism , Deer , Hyaluronic Acid/pharmacology , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
19.
Bull Exp Biol Med ; 177(4): 544-551, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39279005

ABSTRACT

We developed a model of inflammation and airway remodeling in C57 mice provoked by exosomes derived from bone marrow mesenchymal stem cells infected by respiratory syncytial virus (RSV). The mean size of control and infected exosomes in vitro were 167.9 and 118.5 nm, respectively. After induction of modeled pathology, the severity of airway inflammation and its remodeling were analyzed by histopathological methods. In addition, the blood levels of inflammatory factors IL-10, IL-17, transforming growth factor-ß (TGF-ß), and TNFα were assayed; in the lung tissues, the expression levels of MMP-2, MMP-9, α-smooth muscle actin (α-SMA), and TGF-ß were measured. In the developed model, the effects of RSV-induced and non-induced exosomes were compared with those of inactivated and non-inactivated RSV. Intranasal administration of RSV-induced exosomes decreased the levels of serum inflammatory factors IL-10 and IL-17 and increased the expression of serum proinflammatory cytokine TNFα. Increased levels of MMP-2, MMP-9, and α-SMA, enhanced expression of TGF-ß in the lung tissue, and pathological staining of the lung tissues indicated infiltration with inflammatory cells and luminal constriction. Thus, RSV-induced exosomes can provoke airway inflammation and remodeling in mice similar to RSV, while non-induced exosomes cannot produce such alterations.


Subject(s)
Airway Remodeling , Disease Models, Animal , Exosomes , Interleukin-10 , Interleukin-17 , Matrix Metalloproteinase 2 , Mesenchymal Stem Cells , Mice, Inbred C57BL , Respiratory Syncytial Virus Infections , Tumor Necrosis Factor-alpha , Animals , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Matrix Metalloproteinase 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Interleukin-10/metabolism , Interleukin-10/blood , Interleukin-17/metabolism , Lung/pathology , Lung/virology , Lung/metabolism , Matrix Metalloproteinase 9/metabolism , Respiratory Syncytial Viruses/pathogenicity , Transforming Growth Factor beta/metabolism , Actins/metabolism , Inflammation/pathology , Inflammation/metabolism , Bone Marrow Cells/metabolism , Female
20.
Genes Genomics ; 46(10): 1201-1208, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39259486

ABSTRACT

BACKGROUND: Primary ovarian insufficiency (POI) is one of the leading female infertility diseases in which ovarian function stops before the age of 40. Reports that POI is associated with transforming growth factor (TGF)-ß/bone morphogenetic protein (BMP) signaling pathway-associated genes (e.g., TGF-ß, and BMP15) have been continuous since publication that the TGF-ß superfamily acts as important regulators for ovary and placenta function in humans. Mechanistically, the secretion of follicle-stimulating hormone, progesterone, and estrogen is affected by the TGF-ß superfamily in granulosa cells, which are involved in the development of theca cells, oocytes, and granulosa cells. OBJECTIVE: This study aimed to identify the association between genes related to the TGF-ß/BMP signaling pathway and the risk of POI pathogenesis. METHODS: Possible associations between six gene polymorphisms and POI susceptibility were examined in 139 patients with POI and 345 control subjects. RESULTS: Allele combination of TGFBR1 rs334348 G > A and TGFBR3 rs1805110G > A exhibited association with decreased POI risk (adjusted odds ratio [AOR] = 0.165; 95% confidence interval [CI] 0.032-0.847; P = 0.031). Also, TGFBR1 rs1590 G > T and rs334348 G > A and TGFBR3 rs1805110 G > A allele combination exhibited association with decreased POI risk (OR = 0.553; 95% CI 0.374-0.816; P = 0.003). CONCLUSION: This study suggests that polymorphisms in the TGF-ß signaling pathway genes can be useful biomarkers for POI diagnosis and treatment.


Subject(s)
Polymorphism, Single Nucleotide , Primary Ovarian Insufficiency , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction , Transforming Growth Factor beta , Humans , Female , Primary Ovarian Insufficiency/genetics , Adult , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Republic of Korea , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Genetic Predisposition to Disease , Case-Control Studies , Bone Morphogenetic Protein 15/genetics , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Proteoglycans , Receptors, Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL