Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 326
Filter
1.
Mol Cancer ; 23(1): 118, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831405

ABSTRACT

Triple negative breast cancer (TNBC) remains exceptionally challenging to treat. While CDK4/6 inhibitors have revolutionized HR + breast cancer therapy, there is limited understanding of their efficacy in TNBC and meaningful predictors of response and resistance to these drugs remain scarce. We conducted an in vivo genome-wide CRISPR screen using palbociclib as a selection pressure in TNBC. Hits were prioritized using microarray data from a large panel of breast cancer cell lines to identify top palbociclib sensitizers. Our study defines TGFß3 as an actionable determinant of palbociclib sensitivity that potentiates its anti-tumor effects. Mechanistically, we show that chronic palbociclib exposure depletes p21 levels, contributing to acquired resistance, and that TGFß3 treatment can overcome this. This study defines TGFß3 as an actionable biomarker that can be used to improve patient stratification for palbociclib treatment and exploits the synergistic interaction between CDK4/6 and TGFß3 to propose a new combinatorial treatment for TNBC.


Subject(s)
Biomarkers, Tumor , Drug Resistance, Neoplasm , Piperazines , Pyridines , Transforming Growth Factor beta3 , Triple Negative Breast Neoplasms , Humans , Piperazines/pharmacology , Piperazines/therapeutic use , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/drug therapy , Pyridines/pharmacology , Pyridines/therapeutic use , Drug Resistance, Neoplasm/genetics , Female , Biomarkers, Tumor/genetics , Cell Line, Tumor , Mice , Animals , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , CRISPR-Cas Systems , Xenograft Model Antitumor Assays , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic/drug effects
2.
Int J Mol Sci ; 25(10)2024 May 20.
Article in English | MEDLINE | ID: mdl-38791596

ABSTRACT

Ovarian follicular fluid (FF) has a direct impact on oocyte quality, playing key roles in fertilization, implantation, and early embryo development. In our recent study, we found FF thromboxane (TX) to be a novel factor inversely correlated with oocyte maturation and identified thrombin, transforming growth factor ß (TGFß), TNF-α, and follicular granulosa cells (GCs) as possible contributors to FF TX production. Therefore, this study sought to investigate the role of TGFß3 in regulating TX generation in human ovarian follicular GCs. TGFß3 was differentially and significantly present in the FF of large and small follicles obtained from IVF patients with average concentrations of 68.58 ± 12.38 and 112.55 ± 14.82 pg/mL, respectively, and its levels were correlated with oocyte maturity. In an in vitro study, TGFß3 induced TX generation/secretion and the converting enzyme-COX-2 protein/mRNA expression both in human HO23 and primary cultured ovarian follicular GCs. While TGFßRI and Smad2/3 signaling was mainly required for COX-2 induction, ERK1/2 appeared to regulate TX secretion. The participation of Smad2/3 and COX-2 in TGFß3-induced TX generation/secretion could be further supported by the observations that Smad2/3 phosphorylation and nuclear translocation and siRNA knockdown of COX-2 expression compromised TX secretion in GCs challenged with TGFß3. Taken together, the results presented here first demonstrated that FF TGFß3 levels differ significantly in IVF patients' large preovulatory and small mid-antral follicles and are positively associated with oocyte maturation. TGFß3 can provoke TX generation by induction of COX-2 mRNA/protein via a TGFßR-related canonical Smad2/3 signaling pathway, and TX secretion possibly by ERK1/2. These imply that TGFß3 is one of the inducers for yielding FF TX in vivo, which may play a role in folliculogenesis and oocyte maturation.


Subject(s)
Cyclooxygenase 2 , Follicular Fluid , Granulosa Cells , Signal Transduction , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta3 , Humans , Female , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Granulosa Cells/metabolism , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Follicular Fluid/metabolism , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Adult , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Ovarian Follicle/metabolism , Oocytes/metabolism , Cells, Cultured
3.
Nat Commun ; 15(1): 4166, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755146

ABSTRACT

Failure of proper ventricular trabeculation is often associated with congenital heart disease. Support from endocardial cells, including the secretion of extracellular matrix and growth factors is critical for trabeculation. However, it is poorly understood how the secretion of extracellular matrix and growth factors is initiated and regulated by endocardial cells. We find that genetic knockout of histone deacetylase 3 in the endocardium in mice results in early embryo lethality and ventricular hypotrabeculation. Single cell RNA sequencing identifies significant downregulation of extracellular matrix components in histone deacetylase 3 knockout endocardial cells. Secretome from cultured histone deacetylase 3 knockout mouse cardiac endothelial cells lacks transforming growth factor ß3 and shows significantly reduced capacity in stimulating cultured cardiomyocyte proliferation, which is remarkably rescued by transforming growth factor ß3 supplementation. Mechanistically, we identify that histone deacetylase 3 knockout induces transforming growth factor ß3 expression through repressing microRNA-129-5p. Our findings provide insights into the pathogenesis of congenital heart disease and conceptual strategies to promote myocardial regeneration.


Subject(s)
Endocardium , Histone Deacetylases , Mice, Knockout , MicroRNAs , Myocytes, Cardiac , Animals , Endocardium/metabolism , Mice , MicroRNAs/metabolism , MicroRNAs/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Myocytes, Cardiac/metabolism , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Cell Proliferation , Myocardium/metabolism , Endothelial Cells/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Extracellular Matrix/metabolism , Female
4.
Cell Cycle ; 23(5): 555-572, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38695374

ABSTRACT

The study investigates molecular changes in the lumbosacral (L/S) spine's yellow ligamentum flavum during degenerative stenosis, focusing on the role of transforming growth factor beta 1-3 (TGF-ß-1-3). Sixty patients with degenerative stenosis and sixty control participants underwent molecular analysis using real-time quantitative reverse transcription reaction technique (RTqPCR), enzyme-linked immunosorbent assay (ELISA), Western blot, and immunohistochemical analysis (IHC). At the mRNA level, study samples showed reduced expression of TGF-ß-1 and TGF-ß-3, while TGF-ß-2 increased by only 4%. Conversely, at the protein level, the study group exhibited significantly higher concentrations of TGF-ß-1, TGF-ß-2, and TGF-ß-3 compared to controls. On the other hand, at the protein level, a statistically significant higher concentration of TGF-ß-1 was observed (2139.33 pg/mL ± 2593.72 pg/mL vs. 252.45 pg/mL ± 83.89 pg/mL; p < 0.0001), TGF-ß-2 (3104.34 pg/mL ± 1192.74 pg/mL vs. 258.86 pg/mL ± 82.98 pg/mL; p < 0.0001), TGF-ß-3 (512.75 pg/mL ± 107.36 pg/mL vs. 55.06 pg/mL ± 9.83 pg/mL, p < 0.0001) in yellow ligaments obtained from patients of the study group compared to control samples. The study did not establish a significant correlation between TGF-ß-1-3 concentrations and pain severity. The findings suggest that molecular therapy aimed at restoring the normal expression pattern of TGF-ß-1-3 could be a promising strategy for treating degenerative stenosis of the L/S spine. The study underscores the potential therapeutic significance of addressing molecular changes at the TGF-ß isoforms level for better understanding and managing degenerative spinal conditions.


Subject(s)
Protein Isoforms , Spinal Stenosis , Humans , Female , Male , Middle Aged , Protein Isoforms/metabolism , Protein Isoforms/genetics , Spinal Stenosis/metabolism , Spinal Stenosis/pathology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Aged , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/genetics , Ligamentum Flavum/metabolism , Ligamentum Flavum/pathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Adult , Lumbar Vertebrae/metabolism , Lumbar Vertebrae/pathology , Lumbosacral Region/pathology , Case-Control Studies
5.
J Pathol ; 263(3): 338-346, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38594209

ABSTRACT

Necrotising sialometaplasia (NSM) is a non-neoplastic lesion mainly arising in the minor salivary glands of the oral cavity. In the clinical features, NSM shows swelling with or without ulceration, and can mimic a malignant disease such as squamous cell carcinoma. Histopathologically, NSM usually shows the lobular architecture that is observed in the salivary glands. Additionally, acinar infarction and squamous metaplasia of salivary ducts and acini are observable. The aetiology of this lesion remains unknown, although it has a characteristic feature that sometimes requires clinical and histopathological differentiation from malignancy. In this study, we investigated upregulated genes in NSM compared with normal salivary glands, and focused on the TGF-ß3 (TGFB3) gene. The results of the histopathological studies clarified that fibroblasts surrounding the lesion express TGF-ß3. Moreover, in vitro studies using mouse salivary gland organoids revealed that TGF-ß3 suppressed salivary gland cell proliferation and induced squamous metaplasia. We demonstrated a possible aetiology of NSM by concluding that increased TGF-ß3 expression during wound healing or tissue regeneration played a critical role in cell proliferation and metaplasia. © 2024 The Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Proliferation , Fibroblasts , Metaplasia , Salivary Glands , Sialometaplasia, Necrotizing , Transforming Growth Factor beta3 , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Metaplasia/pathology , Animals , Salivary Glands/pathology , Salivary Glands/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Sialometaplasia, Necrotizing/pathology , Sialometaplasia, Necrotizing/metabolism , Sialometaplasia, Necrotizing/genetics , Male , Female , Middle Aged , Adult , Organoids/pathology , Organoids/metabolism
6.
J Dent Res ; 103(5): 546-554, 2024 May.
Article in English | MEDLINE | ID: mdl-38619065

ABSTRACT

The intricate formation of the palate involves a series of complex events, yet its mechanistic basis remains uncertain. To explore major cell populations in the palate and their roles during development, we constructed a spatiotemporal transcription landscape of palatal cells. Palate samples from C57BL/6 J mice at embryonic days 12.5 (E12.5), 14.5 (E14.5), and 16.5 (E16.5) underwent single-cell RNA sequencing (scRNA-seq) to identify distinct cell subsets. In addition, spatial enhanced resolution omics-sequencing (stereo-seq) was used to characterize the spatial distribution of these subsets. Integrating scRNA-seq and stereo-seq with CellTrek annotated mesenchymal and epithelial cellular components of the palate during development. Furthermore, cellular communication networks between these cell subpopulations were analyzed to discover intercellular signaling during palate development. From the analysis of the middle palate, both mesenchymal and epithelial populations were spatially segregated into 3 domains. The middle palate mesenchymal subpopulations were associated with tooth formation, ossification, and tissue remodeling, with initial state cell populations located proximal to the dental lamina. The nasal epithelium of the palatal shelf exhibited richer humoral immune responses than the oral side. Specific enrichment of Tgfß3 and Pthlh signals in the midline epithelial seam at E14.5 suggested a role in epithelial-mesenchymal transition. In summary, this study provides high-resolution transcriptomic information, contributing to a deeper mechanistic understanding of palate biology and pathophysiology.


Subject(s)
Mice, Inbred C57BL , Palate , Animals , Mice , Palate/embryology , Transforming Growth Factor beta3/genetics , Single-Cell Analysis , Epithelial Cells , Sequence Analysis, RNA , Gene Expression Regulation, Developmental , Female
7.
J Cancer Res Ther ; 20(1): 275-280, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38554333

ABSTRACT

CONTEXT: Growth factors and cytokines like transforming growth factor beta (TGF-ß) play a key role in the pathogenesis of oral submucous fibrosis. AIMS: To elucidate the role of Salivary TGF-ß isoforms as a predictive and diagnostic marker for oral submucous fibrosis. SETTINGS AND DESIGN: A total of 30 OSMF and 10 control patients were included in this study, and their clinic-epidemiological data was recorded. METHODOLOGY: The expression of TGF-ß genes-TGF-ß1, TGF-ß2, TGF-ß3-was studied by a real-time polymerase chain reaction in tissue and saliva. Patients were given medicinal intervention for 12 weeks along with jaw-opening exercises. Expression of salivary TGF-ß genes was studied at 12 weeks. STATISTICAL ANALYSIS USED: SPSS software version 20. RESULT: Expression of salivary TGF beta isoforms in OSMF was more than in the control group. There was an increase in salivary TGF-ß1, ß2, ß3 expressions with increasing clinical grades of OSMF and advancing the stage of the disease. Expression of all the TGF beta isoforms was decreased after treatment with statistically significant results. Statistically significant correlations were found between the mean difference of TGF-ß1 and the mean difference between mouth opening and tongue protrusion. CONCLUSION: Salivary TGF-ß isoforms may be used in diagnosis, risk assessment, and screening of the entire population at risk of OSMF after its clinical validation. However, adequate sample size and segmental assessment of the expression of TGF-ß isoforms are needed for further evaluation.


Subject(s)
Oral Submucous Fibrosis , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics , Oral Submucous Fibrosis/diagnosis , Oral Submucous Fibrosis/genetics , Oral Submucous Fibrosis/pathology , Transforming Growth Factor beta3/genetics , Protein Isoforms
8.
Biotechnol Lett ; 46(3): 483-495, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38523201

ABSTRACT

OBJECTIVES: We genetically modified dedifferentiated chondrocytes (DCs) using lentiviral vectors and adenoviral vectors encoding TGF-ß3 (referred to as transgenic groups below) and encapsulated these DCs in the microcavitary hydrogel and investigated the combinational effect on redifferentiation of the genetically manipulated DCs. RESULTS: The Cell Counting Kit-8 data indicated that both transgenic groups exhibited significantly higher cell viability in the first week but inferior cell viability in the subsequent timepoints compared with those of the control group. Real-time polymerase chain reaction and western blot analysis results demonstrated that both transgenic groups had a better effect on redifferentiation to some extent, as evidenced by higher expression levels of chondrogenic genes, suggesting the validity of combination with transgenic DCs and the microcavitary hydrogel on redifferentiation. Although transgenic DCs with adenoviral vectors presented a superior extent of redifferentiation, they also expressed greater levels of the hypertrophic gene type X collagen. It is still worth further exploring how to deliver TGF-ß3 more efficiently and optimizing the appropriate parameters, including concentration and duration. CONCLUSIONS: The results demonstrated the better redifferentiation effect of DCs with the combinational use of transgenic TGF-ß3 and a microcavitary alginate hydrogel and implied that DCs would be alternative seed cells for cartilage tissue engineering due to their easily achieved sufficient cell amounts through multiple passages and great potential to redifferentiate to produce cartilaginous extracellular matrix.


Subject(s)
Cell Differentiation , Chondrocytes , Transforming Growth Factor beta3 , Chondrocytes/cytology , Chondrocytes/metabolism , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/pharmacology , Genetic Vectors/genetics , Hydrogels/chemistry , Animals , Cell Survival , Cells, Cultured , Adenoviridae/genetics , Lentivirus/genetics , Cell Dedifferentiation/genetics , Tissue Engineering/methods
9.
Invest Ophthalmol Vis Sci ; 65(3): 35, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38546583

ABSTRACT

Purpose: To investigate the mechanisms underlying the differential roles of TGFß1 and TGFß3 in accelerating corneal epithelial wound healing (CEWH) in diabetic (DM) corneas, with normoglycemia (NL) corneas as the control. Methods: Two types of diabetic mice, human corneal organ cultures, mouse corneal epithelial progenitor cell lines, and bone marrow-derived macrophages (BMDMs) were employed to assess the effects of TGFß1 and TGFß3 on CEWH, utilizing quantitative PCR, western blotting, ELISA, and whole-mount confocal microscopy. Results: Epithelial debridement led to an increased expression of TGFß1 and TGFß3 in cultured human NL corneas, but only TGFß1 in DM corneas. TGFß1 and TGFß3 inhibition was significantly impeded, but exogenous TGFß1 and, more potently, TGFß3 promoted CEWH in cultured TKE2 cells and in NL and DM C57BL6 mouse corneas. Wounding induced similar levels of p-SMAD2/SMAD3 in NL and DM corneas but weaker ERK1/2, Akt, and EGFR phosphorylation in DM corneas compared to NL corneas. Whereas TGFß1 augmented SMAD2/SMAD3 phosphorylation, TGFß3 preferentially activated ERK, PI3K, and EGFR in healing DM corneas. Furthermore, TGFß1 and TGFß3 differentially regulated the expression of S100a9, PAI-1, uPA/tPA, and CCL3 in healing NL and DM corneas. Finally, TGFß1 induced the expression of M1 macrophage markers iNOS, CD86, and CTGF, whereas TGFß3 promoted the expression of M2 markers CD206 and NGF in BMDMs from db/db or db/+ mice. Conclusions: Hyperglycemia disrupts the balanced expression of TGFß3/TGFß1, resulting in delayed CEWH, including impaired sensory nerve regeneration in the cornea. Supplementing TGFß3 in DM wounds may hold therapeutic potential for accelerating delayed wound healing in diabetic patients.


Subject(s)
Corneal Injuries , Diabetes Mellitus, Experimental , Hyperglycemia , Transforming Growth Factor beta3 , Animals , Humans , Mice , Cornea , ErbB Receptors , Mice, Inbred C57BL , Transforming Growth Factor beta3/genetics
10.
Biol Reprod ; 110(1): 116-129, 2024 Jan 13.
Article in English | MEDLINE | ID: mdl-37801702

ABSTRACT

Ovarian hyperstimulation syndrome (OHSS) is a life-threatening and potentially fatal complication during in vitro fertilization treatment. The levels of transforming growth factor-ß1 (TGF-ß1) are upregulated in human follicular fluid and granulosa-lutein cells (hGL) of OHSS patients and could contribute to the development of OHSS by downregulating steroidogenic acute regulatory protein (StAR) expression. However, whether the same is true for the other two members of the TGF-ß family, TGF-ß2 and -ß3, remains unknown. We showed that all three TGF-ß isoforms were expressed in human follicular fluid. In comparison, TGF-ß1 was expressed at the highest level, followed by TGF-ß2 and TGF-ß3. Compared to non-OHSS patients, follicular fluid levels of TGF-ß1 and TGF-ß3 were significantly upregulated in OHSS patients. The same results were observed in mRNA levels of TGF-ß isoforms in hGL cells and ovaries of OHSS rats. In addition, StAR mRNA levels were upregulated in hGL cells of OHSS patients and the ovaries of OHSS rats. Treatment cells with TGF-ß isoforms downregulated the StAR expression with a comparable effect. Moreover, activations of SMAD3 signaling were required for TGF-ß isoforms-induced downregulation of StAR expression. This study indicates that follicular fluid TGF-ß1 and TGF-ß3 levels could be used as biomarkers and therapeutic targets for the OHSS.


Subject(s)
Ovarian Hyperstimulation Syndrome , Transforming Growth Factor beta1 , Female , Humans , Rats , Animals , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Ovarian Hyperstimulation Syndrome/genetics , RNA, Messenger/metabolism , Protein Isoforms
11.
J Biochem Mol Toxicol ; 38(1): e23581, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38044485

ABSTRACT

Colorectal cancer (CRC) is a common digestive tract tumor with a high incidence and a poor prognosis. Traditional chemotherapy drugs are usually accompanied by unpleasant side effects, highlighting the importance of exploring new adjunctive drugs. In this study, we aimed to explore the role of ursolic acid (UA) in CRC cells. Specifically, HT-29 cells were treated with UA at different concentrations (10, 20, 30, and 40 µM), and the expression of miR-140-5p, tumor growth factor-ß3 (TGF-ß3), ß-catenin, and cyclin D1 was determined by real-time quantitative PCR. The cell cycle and apoptosis were checked by flow cytometry, and cell proliferation was detected by Cell Counting Kit-8 assay. The HT-29 cell model was established through overexpression (miR-140-5p mimics) and interference (miR-140-5p inhibitor) of miR-140-5p. Western blot was used to detect the protein expression of TGF-ß3. We found that UA could inhibit the proliferation of HT-29 cells, block cells in the G1 phase, and promote cell apoptosis. After UA treatment, the expression of miR-140-5p increased and TGF-ß3 decreased. Notably, miR-140-5p downregulated the expression of TGF-ß3, while the overexpression of miR-140-5p exerted a similar function to UA in HT-29 cells. Additionally, the messenger RNA expression of TGF-ß3, ß-catenin, and cyclin D1 was decreased in HT-29 cells after UA treatment. In conclusion, UA inhibited CRC cell proliferation and cell cycle and promoted apoptosis by regulating the miR-140-5p/TGF-ß3 axis, which may be related to the inhibition of Wnt/ß-catenin signaling pathway.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , beta Catenin/metabolism , MicroRNAs/metabolism , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism , Ursolic Acid , Down-Regulation , Cell Proliferation/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic
12.
Medicine (Baltimore) ; 102(49): e36473, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38065913

ABSTRACT

Prostate cancer is the most common malignant tumor of male urogenital system that occurs in prostate epithelium. However, relationship between CAV1 and KRT5 and prostate cancer remains unclear. The prostate cancer datasets GSE114740 and GSE200879 were downloaded from Gene Expression Omnibus generated by GPL11154 and GPL32170. De-batch processing was performed, differentially expressed genes (DEGs) were screened, and weighted gene co-expression network analysis. The construction and analysis of protein-protein interaction network, functional enrichment analysis, gene set enrichment analysis. Gene expression heat map was drawn and immune infiltration analysis was performed. Comparative toxicogenomics database analysis were performed to find the disease most related to core gene. In addition, the cell experiment was performed to verify the role of CAV1 and KRT5 by western blot. Divided into 4 groups: control, prostate cancer, prostate cancer-over expression, and prostate cancer- knock out. TargetScan screened miRNAs that regulated central DEGs; 770 DEGs were identified. According to Gene Ontology analysis, they were mainly concentrated in actin binding and G protein coupled receptor binding. In Kyoto Encyclopedia of Gene and Genome analysis, they were mainly concentrated in PI3K-Akt signal pathway, MAPK signal pathway, and ErbB signal pathway. The intersection of enrichment terms of differentially expressed genes and GOKEGG enrichment terms was mainly concentrated in ErbB signaling pathway and MAPK signaling pathway. Three important modules were generated. The protein-protein interaction network obtained 8 core genes (CAV1, BDNF, TGFB3, FGFR1, PRKCA, DLG4, SNAI2, KRT5). Heat map of gene expression showed that core genes (CAV1, TGFB3, FGFR1, SNAI2, KRT5) are highly expressed in prostate cancer tissues and low in normal tissues. Comparative toxicogenomics database analysis showed that core genes (CAV1, TGFB3, FGFR1, SNAI2, KRT5) were associated with prostate tumor, cancer, tumor metastasis, necrosis, and inflammation. CAV1 and KRT5 are up-regulated in prostate cancer. CAV1 and KRT5 are highly expressed in prostate cancer. The higher expression of CAV1 and KRT5, the worse prognosis.


Subject(s)
Caveolin 1 , Keratin-5 , Prostatic Neoplasms , Transforming Growth Factor beta3 , Humans , Male , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Keratin-5/genetics , Phosphatidylinositol 3-Kinases/genetics , Prostatic Neoplasms/genetics , Transforming Growth Factor beta3/genetics , Caveolin 1/genetics
13.
Sci Rep ; 13(1): 17680, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37848499

ABSTRACT

Despite the prognostic value of arterial stiffness (AS) and pulsatile hemodynamics (PH) for cardiovascular morbidity and mortality, epigenetic modifications that contribute to AS/PH remain unknown. To gain a better understanding of the link between epigenetics (DNA methylation) and AS/PH, we examined the relationship of eight measures of AS/PH with CpG sites and co-methylated regions using multi-ancestry participants from Trans-Omics for Precision Medicine (TOPMed) Multi-Ethnic Study of Atherosclerosis (MESA) with sample sizes ranging from 438 to 874. Epigenome-wide association analysis identified one genome-wide significant CpG (cg20711926-CYP1B1) associated with aortic augmentation index (AIx). Follow-up analyses, including gene set enrichment analysis, expression quantitative trait methylation analysis, and functional enrichment analysis on differentially methylated positions and regions, further prioritized three CpGs and their annotated genes (cg23800023-ETS1, cg08426368-TGFB3, and cg17350632-HLA-DPB1) for AIx. Among these, ETS1 and TGFB3 have been previously prioritized as candidate genes. Furthermore, both ETS1 and HLA-DPB1 have significant tissue correlations between Whole Blood and Aorta in GTEx, which suggests ETS1 and HLA-DPB1 could be potential biomarkers in understanding pathophysiology of AS/PH. Overall, our findings support the possible role of epigenetic regulation via DNA methylation of specific genes associated with AIx as well as identifying potential targets for regulation of AS/PH.


Subject(s)
Atherosclerosis , Epigenesis, Genetic , Humans , Epigenome , Transforming Growth Factor beta3/genetics , Precision Medicine , Genome-Wide Association Study , DNA Methylation , CpG Islands/genetics , Atherosclerosis/genetics
14.
Mol Biol Rep ; 50(7): 6121-6131, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37204543

ABSTRACT

INTRODUCTION: Multiple sclerosis (MS) is an acute demyelinating disease with an autoimmune nature, followed by gradual neurodegeneration and enervating scar formation. Dysregulated immune response is a crucial dilemma contributing to the pathogenesis of MS. The role of chemokines and cytokines, such as transforming growth factor-ß (TGF-ß), have been recently highlighted regarding their altered expressions in MS. TGF-ß has three isoforms, TGF-ß1, TGF-ß2, and TGF-ß3, that are structurally similar; however, they can show different functions. RESULTS: All three isoforms are known to induce immune tolerance by modifying Foxp3+ regulatory T cells. Nevertheless, there are controversial reports concerning the role of TGF-ß1 and 2 in the progression of scar formation in MS. At the same time, these proteins also improve oligodendrocyte differentiation and have shown neuroprotective behavior, two cellular processes that suppress the pathogenesis of MS. TGF-ß3 shares the same properties but is less likely contributes to scar formation, and its direct role in MS remains elusive. DISCUSSION: To develop novel neuroimmunological treatment strategies for MS, the optimal strategy could be the one that causes immune modulation, induces neurogenesis, stimulates remyelination, and prevents excessive scar formation. Therefore, regarding its immunological properties, TGF-ß could be an appropriate candidate; however, contradictory results of previous studies have questioned its role and therapeutic potential in MS. In this review article, we provide an overview of the role of TGF-ß in immunopathogenesis of MS, related clinical and animal studies, and the treatment potential of TGF-ß in MS, emphasizing the role of different TGF-ß isoforms.


Subject(s)
Multiple Sclerosis , Transforming Growth Factor beta , Animals , Cicatrix , Multiple Sclerosis/genetics , Protein Isoforms/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/genetics , Humans
15.
Oral Surg Oral Med Oral Pathol Oral Radiol ; 136(2): e109-e115, 2023 08.
Article in English | MEDLINE | ID: mdl-37246056

ABSTRACT

OBJECTIVE: Coronoid process hyperplasia (CPH) of the mandible can lead to restricted mouth opening and maxillofacial deformities, which have been hypothesized to be closely associated with genetics. This study investigated the relationship between congenital CPH and TGFB3 mutation in a family of patients with CPH. STUDY DESIGN: A limited mouth opening proband with CPH underwent whole-exome gene sequencing in November 2019, and the results confirmed compound heterozygous mutations in the TGFB3 gene. Subsequently, clinical imaging and genetic testing were performed on 10 other individuals in his family. RESULTS: A total of 9 people in this family have CPH. Among them, 6 have the same exon compound heterozygous mutation sites of the TGFB3 gene (chr14-76446905 and chr14-76429713), accompanied by homozygous or heterozygous mutations in the 3'untranslated region (3'UTR) of the TGFB3 gene (chr14:76429555). The other 3 individuals have a homozygous mutation in the 3'untranslated region of the TGFB3 gene. CONCLUSION: The heterogeneous compound mutation of the TGFB3 gene or the homozygous mutation of 3'UTR of the TGFB3 gene may be correlated with CPH. In addition, the specifically related mechanism needs to be confirmed by further genetic animal experiments.


Subject(s)
Mandible , Transforming Growth Factor beta3 , Humans , Transforming Growth Factor beta3/genetics , Hyperplasia/genetics , Hyperplasia/pathology , 3' Untranslated Regions , Mutation/genetics , Mandible/pathology
16.
Ann Clin Lab Sci ; 52(6): 976-985, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36564064

ABSTRACT

OBJECTIVE: Chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) plays an important role in osteoarthritis (OA) treatment. Studies have reported the association of transforming growth factor beta-3 (TGF-ß3) with chondrogenic differentiation of BMSCs. In this study, the upstream mechanism and functions of TGF-ß3 in chondrogenic differentiation of BMSCs were explored. METHODS AND RESULTS: Flow cytometry was performed for the positive and negative MSC markers. Chondrogenic differentiation of BMSCs was evaluated by Alcian blue staining. Gene expression and protein expression levels were measured by RT-qPCR and western blotting, respectively. Relationship between let-7a-3p and TGF-ß3 was confirmed using bioinformatics analysis, luciferase reporter and RNA pulldown assays. Subcellular distributions of TGF-ß3 and let-7a-3p were determined by FISH. In this study, BMSCs were identified to possess the characteristics of mesenchymal stem cells. TGF-ß3 was found to induce chondrogenic differentiation of BMSCs. Mechanically, TGF-ß3 was verified to be targeted by let-7a-3p. In rescue assays, let-7a-3p overexpression reversed the effects of TGF-ß3 overexpression on chondrogenic differentiation of BMSCs. CONCLUSION: Let-7a-3p inhibits chondrogenic differentiation of bone marrow mesenchymal stem cells by targeting TGF-ß3.


Subject(s)
Mesenchymal Stem Cells , Transforming Growth Factor beta3 , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/pharmacology , Transforming Growth Factor beta3/metabolism , Cell Differentiation/genetics , Blotting, Western , Chondrogenesis/genetics , Cells, Cultured , Bone Marrow Cells/metabolism
17.
BMC Womens Health ; 22(1): 543, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36564776

ABSTRACT

BACKGROUND: Abnormal uterine bleeding (AUB) is irregular menstrual bleeding which has great impact on female health and life style. Various genetic factors are involved in etiology and pathology of AUB. Present study was designed to explore the association of PTGFR, MMP9, MMP2, TGFB3 and VEGFB with AUB. METHODS: Blood samples of 212 females with AUB were collected along with age-matched healthy control. Expression variation of targeted genes was evaluated using qPCR. Present study cohort was divided into different groups based on demographic parameters and all targeted genes were correlated with study demographics. RESULTS: Expression of targeted genes was significantly (P < 0.001) downregulated in females with AUB compared to control. Reduced (P < 0.01) expression of targeted genes was observed in all age groups (21-30, 31-40, 41-50 year) of AUB patients compared to respective control. Expression of VEGFB increased (P < 0.05) in AUB females with > 9 days bleeding compared to AUB patient had < 9 days bleeding. AUB women with miscarriage history showed upregulation in MMP2, TGFB3 (P < 0.05), and downregulation in MMP9 and VEGFB (P < 0.05) expression compared to AUB group with no miscarriage history. Expression of MMP2 increased (P < 0.05) in AUB females with > 60 kg body weigh compared to AUB patient with < 60 kg weight. CONCLUSION: Present study open a new window for diagnosis of AUB at early stages and suggested a possible involvement of PTGFR, MMP9, MMP2, TGFB3 and VEGFB as candidate biomarkers in AUB.


Subject(s)
Matrix Metalloproteinase 2 , Uterine Hemorrhage , Female , Humans , Uterine Hemorrhage/genetics , Uterine Hemorrhage/diagnosis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Transforming Growth Factor beta3/genetics , Pakistan , Vasoconstriction
18.
Theranostics ; 12(12): 5470-5487, 2022.
Article in English | MEDLINE | ID: mdl-35910794

ABSTRACT

Rationale: Wound healing is among the most complicated physiological processes and requires the synchronization of various cell types with distinct roles to re-establish the condition of the original skin. Patients affected by peripheral neuropathies often experience failure to heal. Loss of Schwann cells (SCs), a crucial population of peripheral nervous system cells in skin, may contribute to chronic wounds. However, the role of SCs in wound healing are poorly understood. Methods: The activity of SCs was investigated by using a cell atlas of the wound healing process, which was generated by integrating single-cell RNA sequencing (scRNA-seq) libraries covering different states of mouse back skin. The results of in silico analysis were validated by in vitro cell culture and in vivo mouse model. Selective inhibitors and conditional RNAi by virus transfection were utilized to investigate the role of SCs in wound healing. Findings from mouse experiments were further verified in scRNA-seq analysis of diabetic patients. Results: Our in silico analysis revealed the heterogeneous cellular components of skin and the dynamic interactions of neural crest derived cells (NCs) with other cell types. We found that SCs dedifferentiated at an early stage of wound repair with upregulated Wnt signaling. We also identified dedifferentiated SC (dSC) defect in diabetic wounds in both mouse and human. Wnt inhibition at the wound site repressed SC dedifferentiation, leading to defective repair. Furthermore, dSCs derived TGF-ß3, which is context-dependent, promoted the migration of fibroblasts and keratinocytes. Moreover, TGF-ß3 supplementation enhanced the healing of chronic wounds in diabetic mice with impaired SCs. Conclusion: Our study thus advances the understanding of the roles of neural-derived cells in skin regeneration and suggests a potential therapeutic strategy for wound healing disorders.


Subject(s)
Cell Dedifferentiation , Diabetes Mellitus, Experimental , Peripheral Nervous System Diseases , Schwann Cells , Transforming Growth Factor beta3 , Wound Healing , Animals , Cell Dedifferentiation/genetics , Cell Dedifferentiation/physiology , Humans , Mice , Peripheral Nervous System Diseases/genetics , Schwann Cells/physiology , Skin/injuries , Skin/innervation , Transforming Growth Factor beta3/genetics , Wound Healing/genetics , Wound Healing/physiology
19.
Front Immunol ; 13: 746360, 2022.
Article in English | MEDLINE | ID: mdl-35774789

ABSTRACT

Introduction: TGF-ß and its receptors play a crucial role in asthma pathogenesis and bronchial remodeling in the course of the disease. TGF-ß1, TGF-ß2, and TGF-ß3 isoforms are responsible for chronic inflammation, bronchial hyperreactivity, myofibroblast activation, fibrosis, bronchial remodeling, and change the expression of approximately 1000 genes in asthma. TGF-ß SNPs are associated with the elevated plasma level of TGF-ß1, an increased level of total IgE, and an increased risk of remodeling of bronchi. Methods: The analysis of selected TGF-ß1, TGF-ß2, TGF-ß3-related single-nucleotide polymorphisms (SNP) was conducted on 652 DNA samples with an application of the MassARRAY® using the mass spectrometry (MALDI-TOF MS). Dataset was randomly split into training (80%) and validation sets (20%). For both asthma diagnosis and severity prediction, the C5.0 modelling with hyperparameter optimization was conducted on: clinical and SNP data (Clinical+TGF), only clinical (OnlyClinical) and minimum redundancy feature selection set (MRMR). Area under ROC (AUCROC) curves were compared using DeLong's test. Results: Minor allele carriers (MACs) in SNP rs2009112 [OR=1.85 (95%CI:1.11-3.1), p=0.016], rs2796821 [OR=1.72 (95%CI:1.1-2.69), p=0.017] and rs2796822 [OR=1.71 (95%CI:1.07-2.71), p=0.022] demonstrated an increased odds of severe asthma. Clinical+TGF model presented better diagnostic potential than OnlyClinical model in both training (p=0.0009) and validation (AUCROC=0.87 vs. 0.80,p=0.0052). At the same time, the MRMR model was not worse than the Clinical+TGF model (p=0.3607 on the training set, p=0.1590 on the validation set), while it was better in comparison with the Only Clinical model (p=0.0010 on the training set, p=0.0235 on validation set, AUCROC=0.85 vs. 0.87). On validation set Clinical+TGF model allowed for asthma diagnosis prediction with 88.4% sensitivity and 73.8% specificity. Discussion: Derived predictive models suggest the analysis of selected SNPs in TGF-ß genes in combination with clinical factors could predict asthma diagnosis with high sensitivity and specificity, however, the benefit of SNP analysis in severity prediction was not shown.


Subject(s)
Asthma , Transforming Growth Factor beta1 , Asthma/diagnosis , Asthma/genetics , Case-Control Studies , Cytokines/genetics , Data Mining , Humans , Polymorphism, Single Nucleotide , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta2 , Transforming Growth Factor beta3/genetics
20.
F S Sci ; 3(3): 288-298, 2022 08.
Article in English | MEDLINE | ID: mdl-35643626

ABSTRACT

OBJECTIVE: To clarify whether a mediator complex subunit 12 (MED12) gain-of-function mutation induces leiomyoma cell properties in human uterine smooth muscle cells (USMCs). DESIGN: Experimental study. SETTING: Academic research laboratory. PATIENT(S): Women undergoing hysterectomy for leiomyoma. INTERVENTION(S): CRISPR/Cas9-mediated genome editing to introduce an MED12 gain-of-function mutation (G44D) into human USMCs. MAIN OUTCOME MEASURE(S): Cell proliferation, collagen production, and in vivo tumorigenicity of USMCs with vs. without the MED12 mutation. RESULT(S): Uterine smooth muscle cells isolated from the uterine myometrium of a 44-year-old patient were subjected to lentiviral vector-mediated gene transduction of the fluorescent protein Venus, followed by long-term passage. Uterine smooth muscle cells with a normal female karyotype, high cell proliferative activity, and Venus expression, but without stem/progenitor cell populations, were obtained and designated as USMC44. Using CRISPR/Cas9-mediated genome editing, mtUSMC44 (MED12, 131G>A, p.G44D) and mock USMC44 without MED12 mutation (wtUSMC44) were established from USMC44. wtUSMC44 and mtUSMC44 showed similar cell proliferation activity, even in the presence of estradiol and progesterone (EP) together with transforming growth factor-beta 3 (TGFB3). In addition, wtUSMC44 and mtUSMC44 generated similar tiny smooth muscle-like tissue constructs when xenotransplanted beneath the kidney capsule in immunodeficient mice treated with EP alone or TGFB3. In contrast, mtUSMC44 produced more collagen type I than wtUSMC in vitro, and this production was likely enhanced by EP and TGFB3. CONCLUSION(S): The results suggest that the MED12 gain-of-function mutation is involved in collagen production. Although approximately 70% of leiomyomas have MED12 mutations, additional factors and/or events other than MED12 and/or myometrial stem/progenitor cells may be required for fully inducing leiomyoma cell properties, including transformation, in USMCs.


Subject(s)
Leiomyoma , Uterine Neoplasms , Adult , Animals , Female , Gain of Function Mutation , Humans , Leiomyoma/genetics , Mediator Complex/genetics , Mice , Mutation , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta3/genetics , Uterine Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...