Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
J Dent Res ; 103(5): 546-554, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38619065

RESUMO

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.


Assuntos
Camundongos Endogâmicos C57BL , Palato , Animais , Camundongos , Palato/embriologia , Fator de Crescimento Transformador beta3/genética , Análise de Célula Única , Células Epiteliais , Análise de Sequência de RNA , Regulação da Expressão Gênica no Desenvolvimento , Feminino
2.
J Cancer Res Ther ; 20(1): 275-280, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38554333

RESUMO

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.


Assuntos
Fibrose Oral Submucosa , Fator de Crescimento Transformador beta , Humanos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/genética , Fibrose Oral Submucosa/diagnóstico , Fibrose Oral Submucosa/genética , Fibrose Oral Submucosa/patologia , Fator de Crescimento Transformador beta3/genética , Isoformas de Proteínas
3.
Invest Ophthalmol Vis Sci ; 65(3): 35, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38546583

RESUMO

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.


Assuntos
Lesões da Córnea , Diabetes Mellitus Experimental , Hiperglicemia , Fator de Crescimento Transformador beta3 , Animais , Humanos , Camundongos , Córnea , Receptores ErbB , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta3/genética
4.
Biotechnol Lett ; 46(3): 483-495, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38523201

RESUMO

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.


Assuntos
Diferenciação Celular , Condrócitos , Fator de Crescimento Transformador beta3 , Condrócitos/citologia , Condrócitos/metabolismo , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/farmacologia , Vetores Genéticos/genética , Hidrogéis/química , Animais , Sobrevivência Celular , Células Cultivadas , Adenoviridae/genética , Lentivirus/genética , Desdiferenciação Celular/genética , Engenharia Tecidual/métodos
5.
J Biochem Mol Toxicol ; 38(1): e23581, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38044485

RESUMO

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.


Assuntos
Neoplasias Colorretais , MicroRNAs , Humanos , beta Catenina/metabolismo , MicroRNAs/metabolismo , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo , Ácido Ursólico , Regulação para Baixo , Proliferação de Células/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica
6.
Biol Reprod ; 110(1): 116-129, 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-37801702

RESUMO

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.


Assuntos
Síndrome de Hiperestimulação Ovariana , Fator de Crescimento Transformador beta1 , Feminino , Humanos , Ratos , Animais , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/metabolismo , Síndrome de Hiperestimulação Ovariana/genética , RNA Mensageiro/metabolismo , Isoformas de Proteínas
7.
Medicine (Baltimore) ; 102(49): e36473, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38065913

RESUMO

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.


Assuntos
Caveolina 1 , Queratina-5 , Neoplasias da Próstata , Fator de Crescimento Transformador beta3 , Humanos , Masculino , Biologia Computacional , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Queratina-5/genética , Fosfatidilinositol 3-Quinases/genética , Neoplasias da Próstata/genética , Fator de Crescimento Transformador beta3/genética , Caveolina 1/genética
8.
Oral Surg Oral Med Oral Pathol Oral Radiol ; 136(2): e109-e115, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37246056

RESUMO

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.


Assuntos
Mandíbula , Fator de Crescimento Transformador beta3 , Humanos , Fator de Crescimento Transformador beta3/genética , Hiperplasia/genética , Hiperplasia/patologia , Regiões 3' não Traduzidas , Mutação/genética , Mandíbula/patologia
9.
Ann Clin Lab Sci ; 52(6): 976-985, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36564064

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais , Fator de Crescimento Transformador beta3 , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/farmacologia , Fator de Crescimento Transformador beta3/metabolismo , Diferenciação Celular/genética , Western Blotting , Condrogênese/genética , Células Cultivadas , Células da Medula Óssea/metabolismo
10.
Front Immunol ; 13: 746360, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35774789

RESUMO

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.


Assuntos
Asma , Fator de Crescimento Transformador beta1 , Asma/diagnóstico , Asma/genética , Estudos de Casos e Controles , Citocinas/genética , Mineração de Dados , Humanos , Polimorfismo de Nucleotídeo Único , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta2 , Fator de Crescimento Transformador beta3/genética
11.
F S Sci ; 3(3): 288-298, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35643626

RESUMO

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.


Assuntos
Leiomioma , Neoplasias Uterinas , Adulto , Animais , Feminino , Mutação com Ganho de Função , Humanos , Leiomioma/genética , Complexo Mediador/genética , Camundongos , Mutação , Miócitos de Músculo Liso/metabolismo , Fator de Crescimento Transformador beta3/genética , Neoplasias Uterinas/genética
12.
Stem Cell Rev Rep ; 18(6): 2045-2058, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35303271

RESUMO

BACKGROUND: Pressure ulcers (PUs), a result of ischemic reperfusion (IR) injuries, are prevalent skin problems which show refractoriness against standard therapeutic approaches. Besides, scar formation is a critical complication of ulcers that affects functionality and the skin's cosmetic aspect. The current study aimed to investigate the effects of placenta-derived human amniotic epithelial cells (hAECs), as important agents of regenerative medicine and stem cell therapy, on accelerating the healing of IR ulcers in mice. We also evaluated the effects of these cells on reducing the TGFß-induced scar formation. METHODS: Male Balb/c mice at the age of 6-8 weeks were subjected to three IR cycles. Afterward, the mice were divided into three experimental groups (n = 6 per group), including the control group, vehicle group, and hAECs treatment group. Mice of the treatment group received 100 µL of fresh hAECs 1 × 106 cell/ml suspension in PBS. Afterward, mice were assessed by histological, stereological, molecular, and western blotting techniques at 3, 7, 14, and 21 days after wounding. RESULTS: The histological and stereological results showed the most diminutive scar formation and better healing in the hAECs treated group compared to control group. Furthermore, our results demonstrated that the expression level of Col1A1 on days 3, 14, and 21 in the hAECs treated group was significantly lower than control. Additionally, injection of hAECs significantly reduced the expression level of Col3A1 on days 3, 7, and 21 while increased Col3A1 on the day 14. Otherwise, in the hAECs treated group, the expression levels of VEGFA on days 7 and 14 were higher, which showed that hAECs could promote angiogenesis and wound healing. Also, cell therapy significantly lowered the protein levels of TGF-ß1 on day 14, while the protein level of TGF-ß3 on day 14 was significantly higher. This data could demonstrate the role of hAECs in scar reduction in IR wounds. CONCLUSION: These results suggest that hAECs can promote re-epithelialization and wound closure in an animal model of PU. They also reduced scar formation during wound healing by reducing the expression of TGF-ß1/ TGF-ß3 ratio.


Assuntos
Cicatriz , Células Epiteliais , Traumatismo por Reperfusão , Cicatrização , Âmnio/citologia , Animais , Cicatriz/terapia , Células Epiteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Placenta/citologia , Gravidez , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/terapia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo , Úlcera/metabolismo
13.
Reprod Sci ; 29(8): 2401-2413, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35028925

RESUMO

Maternal exposure to dibutyl phthalate (DBP) may result in ovarian dysfunction in female offspring. However, the underlying mechanisms remain elusive. Pregnant Sprague-Dawley rats were intraperitoneally injected with different doses of DBP, estradiol, and corn oil from gestational day 7 until the end of lactation. The reproductive characteristics, mRNA, and protein expression of ovaries for the adult female offspring were compared. KGN cells were cultured in vitro with DBP, estrogen receptor antagonist, or ALK-5 inhibitor. Genes, proteins, estradiol, and progesterone expressed by KGN, cell proliferation, and apoptosis were measured respectively. Maternal perinatal exposure to DBP induced prolonged estrous period, increased secondary follicles, significant decreased mRNA, and protein levels of TGF-ß2, TGF-ß3, and TGF-ßRII in ovaries of the adult female offspring, but none difference for serum levels of sex hormones, ovarian TGF-ß1, and estrogen receptor. The mRNA levels of LHR, FSHR, and CYP19a in ovaries were also decreased. DBP might decrease the mRNA of TGF-ß2, TGF-ß3, and TGF-ßR II of KGN. DBP can inhibit the mRNA of CYP19 at 24 h, which might be blocked by the estrogen receptor antagonist, whose effects were attenuated at 48 h. DBP combined with FSH might time-dependently regulate the gene expression of TGF-ßR II, inhibitory at 24 h, but stimulative at 48 h, which could be blocked by the ALK5 inhibitor. However, the protein expressed by KGN was not influenced by DBP. DBP stimulated the proliferation of KGN at 24 h, which could be blocked by estrogen receptor antagonist, but attenuated at 48 h. The progesterone in culture medium secreted by KGN was decreased by DBP at 24 h. Maternal perinatal exposure to DBP induced decreased gene expression of TGF-ß signaling and functional proteins in ovaries of the adult female offspring. Molecular cross-talk between estrogen receptor and TGF-ß signaling pathway may play role in the mechanism of granulosa dysfunction induced by DBP.


Assuntos
Dibutilftalato , Exposição Materna , Ovário , Efeitos Tardios da Exposição Pré-Natal , Animais , Dibutilftalato/toxicidade , Regulação para Baixo , Estradiol , Antagonistas do Receptor de Estrogênio , Feminino , Exposição Materna/efeitos adversos , Ovário/fisiopatologia , Gravidez , Progesterona , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/metabolismo , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo
14.
Exp Cell Res ; 410(1): 112945, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34838812

RESUMO

OBJECTIVE: Mesenchymal stem cells (MSCs), especially genetically modified MSCs, have become a promising therapeutic approach for the treatment of rheumatoid arthritis (RA) through modulating immune responses. However, most MSCs used in the treatment of RA are modified based on a single gene. In this study, we evaluated the therapeutic effects of human BMSCs (hBMSCs) with COX-2 silence and TGF-ß3 overexpression in the treatment of RA in a rabbit model. MATERIALS AND METHODS: hBMSCs were cotransfected with shCOX-2 and TGF-ß3 through lentiviral vector delivery. After SPIO-Molday ION Rhodamine-B™ (MIRB) labeling, lenti-shCOX2-TGF-ß3 hBMSCs, lenti-shCOX2 hBMSCs, lenti-TGF-ß3 hBMSCs, hBMSCs without genetic modification, or phosphate-buffered saline (PBS) were injected into the knee joint of rabbits with antigen-induced arthritis (AIA). The diameter of the knee joint and soft-tissue swelling score (STS) were recorded, and the levels of inflammatory mediators, including interleukin-1ß (IL-1ß), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and prostaglandin E2 (PGE2) were evaluated by ELISA. Clinical 3.0T MR imaging (MRI) was used to track the distribution and dynamic migration of hBMSCs in the joint. Histopathological and immunohistochemical assays were conducted to localize labeled hBMSCs and assess the alteration of synovial hyperplasia, inflammatory cell infiltration, and cartilage damage. RESULTS: COX-2 silencing and TGF-ß3 overexpression in hBMSCs were confirmed through real-time PCR and Western blot analyses. Reduced joint diameter, soft-tissue swelling (STS) score, and PGE2, IL-1ß, and TNF-α expression were detected 4 weeks after injection of MIRB-labeled lenti-shCOX2-TGF-ß3 hBMSCs into the joint in rabbits with AIA. Eight weeks after hBMSC injection, reduced inflammatory cell infiltration, improved hyperplasia of the synovial lining, recovered cartilage damage, and increased matrix staining were observed in joints injected with lenti-shCOX2-TGF-ß3 hBMSCs and lenti-shCOX2 hBMSCs. Slight synovial hyperplasia, no surface fibrillation, and strong positive expression of collagen II staining in chondrocytes and cartilage matrix were detected in the joints 12 weeks after injection of lenti-shCOX2-TGF-ß3 hBMSCs. In addition, hBMSCs were detected by MRI imaging throughout the process of hBMSC treatment. CONCLUSION: Intra-articular injection of hBMSCs with COX-2 silence and TGFß3 overexpression not only significantly inhibited joint inflammation and synovium hyperplasia, but also protected articular cartilage at the early stage. In addition, intra-articular injection of hBMSCs with COX-2 silence and TGFß3 overexpression promoted chondrocyte and matrix proliferation. This study provides an alternative therapeutic strategy for the treatment of RA using genetically modified hBMSCs.


Assuntos
Artrite Reumatoide/genética , Ciclo-Oxigenase 2/genética , Inflamação/genética , Fator de Crescimento Transformador beta3/genética , Animais , Antígenos/farmacologia , Artrite Reumatoide/etiologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Condrócitos/imunologia , Condrócitos/metabolismo , Ciclo-Oxigenase 2/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Modelos Animais de Doenças , Humanos , Imunidade/genética , Inflamação/etiologia , Inflamação/imunologia , Inflamação/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Coelhos
15.
Sci Rep ; 11(1): 22365, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34785671

RESUMO

Antibody function is typically entirely dictated by the Complementarity Determining Regions (CDRs) that directly bind to the antigen, while the framework region acts as a scaffold for the CDRs and maintains overall structure of the variable domain. We recently reported that the rabbit monoclonal antibody 4A11 (rbt4A11) disrupts signaling through both TGFß2 and TGFß3 (Sun et al. in Sci Transl Med, 2021. https://doi.org/10.1126/scitranslmed.abe0407 ). Here, we report a dramatic, unexpected discovery during the humanization of rbt4A11 where, two variants of humanized 4A11 (h4A11), v2 and v7 had identical CDRs, maintained high affinity binding to TGFß2/3, yet exhibited distinct differences in activity. While h4A11.v7 completely inhibited TGFß2/3 signaling like rbt4A11, h4A11.v2 did not. We solved crystal structures of TGFß2 complexed with Fab fragments of h4A11.v2 or h4A11.v7 and identified a novel interaction between the two heavy chain molecules in the 2:2 TGFb2:h4A11.v2-Fab complex. Further characterization revealed that framework residue variations at either position 19, 79 or 81 (Kabat numbering) of the heavy chain strikingly converts h4A11.v2 into an inhibitory antibody. Our work suggests that in addition to CDRs, framework residues and interactions between Fabs in an antibody could be engineered to further modulate activity of antibodies.


Assuntos
Substituição de Aminoácidos , Anticorpos Monoclonais Humanizados/química , Fragmentos Fab das Imunoglobulinas/química , Região Variável de Imunoglobulina/química , Fator de Crescimento Transformador beta2/química , Fator de Crescimento Transformador beta3/química , Animais , Anticorpos Monoclonais Humanizados/genética , Cristalografia por Raios X , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Estrutura Quaternária de Proteína , Coelhos , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta3/genética
16.
Carcinogenesis ; 42(7): 913-923, 2021 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-34057989

RESUMO

Chordoma is a rare bone tumor arising from notochordal remnants, but the underlying mechanism remains elusive. By integrated mRNA and microRNA analyses, we found significant downregulation of TGFB3 along with upregulation of its inhibitor, miR-29 family in chordoma comparing with notochord. Somatic copy number gains of miR-29 loci in chordoma highlighted a mechanism of inactivation of TGFB3 signaling in tumor formation. In zebrafish, knockout and knockdown homologous tgfb3 resulted in a chordoma-like neoplasm. On the other hand, Smad7 negative feedback regulation of transforming growth factor-ß (TGF-ß) signaling is retentive in chordoma cell UM-Chor1 despite its disruption in most cancer cells (e.g. A549). Therefore, contrary to other cancers, exogenous TGF-ß activated Smad7 by downregulating miR-182 and inhibited cell migration and invasion in UM-Chor1. Meanwhile, TGF-ß decreased chordoma characteristic protein Brachyury. Altogether, downregulation of TGFB3 causes chordomagenesis, showing a feasible target for therapies. The retention of Smad7 negative regulation may maintain the suppressor role of TGF-ß in chordoma.


Assuntos
Biomarcadores Tumorais/metabolismo , Cordoma/patologia , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta3/antagonistas & inibidores , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Cordoma/genética , Cordoma/metabolismo , Humanos , Prognóstico , Proteína Smad7/genética , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo , Células Tumorais Cultivadas
17.
Stem Cell Res Ther ; 12(1): 251, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33926568

RESUMO

BACKGROUND: Due to its low capacity for self-repair, articular cartilage is highly susceptible to damage and deterioration, which leads to the development of degenerative joint diseases such as osteoarthritis (OA). Menstrual blood-derived mesenchymal stem/stromal cells (MenSCs) are much less characterized, as compared to bone marrow mesenchymal stem/stromal cells (BMMSCs). However, MenSCs seem an attractive alternative to classical BMMSCs due to ease of access and broader differentiation capacity. The aim of this study was to evaluate chondrogenic differentiation potential of MenSCs and BMMSCs stimulated with transforming growth factor ß (TGF-ß3) and activin A. METHODS: MenSCs (n = 6) and BMMSCs (n = 5) were isolated from different healthy donors. Expression of cell surface markers CD90, CD73, CD105, CD44, CD45, CD14, CD36, CD55, CD54, CD63, CD106, CD34, CD10, and Notch1 was analyzed by flow cytometry. Cell proliferation capacity was determined using CCK-8 proliferation kit and cell migration ability was evaluated by scratch assay. Adipogenic differentiation capacity was evaluated according to Oil-Red staining and osteogenic differentiation according to Alizarin Red staining. Chondrogenic differentiation (activin A and TGF-ß3 stimulation) was investigated in vitro and in vivo (subcutaneous scaffolds in nude BALB/c mice) by expression of chondrogenic genes (collagen type II, aggrecan), GAG assay and histologically. Activin A protein production was evaluated by ELISA during chondrogenic differentiation in monolayer culture. RESULTS: MenSCs exhibited a higher proliferation rate, as compared to BMMSCs, and a different expression profile of several cell surface markers. Activin A stimulated collagen type II gene expression and glycosaminoglycan synthesis in TGF-ß3 treated MenSCs but not in BMMSCs, both in vitro and in vivo, although the effects of TGF-ß3 alone were more pronounced in BMMSCs in vitro. CONCLUSION: These data suggest that activin A exerts differential effects on the induction of chondrogenic differentiation in MenSCs vs. BMMSCs, which implies that different mechanisms of chondrogenic regulation are activated in these cells. Following further optimization of differentiation protocols and the choice of growth factors, potentially including activin A, MenSCs may turn out to be a promising population of stem cells for the development of cell-based therapies with the capacity to stimulate cartilage repair and regeneration in OA and related osteoarticular disorders.


Assuntos
Células-Tronco Mesenquimais , Ativinas , Animais , Células da Medula Óssea , Diferenciação Celular , Células Cultivadas , Condrogênese , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Osteogênese , Fenótipo , Fator de Crescimento Transformador beta3/genética
18.
PLoS Genet ; 17(4): e1009515, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33914736

RESUMO

Zebrafish exhibit robust regeneration following spinal cord injury, promoted by macrophages that control post-injury inflammation. However, the mechanistic basis of how macrophages regulate regeneration is poorly understood. To address this gap in understanding, we conducted a rapid in vivo phenotypic screen for macrophage-related genes that promote regeneration after spinal injury. We used acute injection of synthetic RNA Oligo CRISPR guide RNAs (sCrRNAs) that were pre-screened for high activity in vivo. Pre-screening of over 350 sCrRNAs allowed us to rapidly identify highly active sCrRNAs (up to half, abbreviated as haCRs) and to effectively target 30 potentially macrophage-related genes. Disruption of 10 of these genes impaired axonal regeneration following spinal cord injury. We selected 5 genes for further analysis and generated stable mutants using haCRs. Four of these mutants (tgfb1a, tgfb3, tnfa, sparc) retained the acute haCR phenotype, validating the approach. Mechanistically, tgfb1a haCR-injected and stable mutant zebrafish fail to resolve post-injury inflammation, indicated by prolonged presence of neutrophils and increased levels of il1b expression. Inhibition of Il-1ß rescues the impaired axon regeneration in the tgfb1a mutant. Hence, our rapid and scalable screening approach has identified functional regulators of spinal cord regeneration, but can be applied to any biological function of interest.


Assuntos
RNA Guia de Cinetoplastídeos/genética , Regeneração/genética , Regeneração da Medula Espinal/genética , Fator de Crescimento Transformador beta1/genética , Proteínas de Peixe-Zebra/genética , Animais , Axônios/metabolismo , Axônios/fisiologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Modelos Animais de Doenças , Macrófagos/metabolismo , Osteonectina/genética , Recuperação de Função Fisiológica/genética , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/patologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Regeneração da Medula Espinal/fisiologia , Fator de Crescimento Transformador beta3/genética , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
19.
BMC Genom Data ; 22(1): 13, 2021 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-33743603

RESUMO

BACKGROUND: Bioengineering has demonstrated the potential of utilising mesenchymal stem cells (MSCs), growth factors, and mechanical stimuli to treat cartilage defects. However, the underlying genes and pathways are largely unclear. This is the first study on screening and identifying the hub genes involved in mechanically enhanced chondrogenesis and their potential molecular mechanisms. METHODS: The datasets were downloaded from the Gene Expression Omnibus (GEO) database and contain six transforming growth factor-beta-3 (TGF-ß3) induced bovine bone marrow-derived MSCs specimens and six TGF-ß3/dynamic-compression-induced specimens at day 42. Screening differentially expressed genes (DEGs) was performed and then analysed via bioinformatics methods. The Database for Annotation, Visualisation, and Integrated Discovery (DAVID) online analysis was utilised to obtain the Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment. The protein-protein interaction (PPI) network of the DEGs was constructed based on data from the STRING database and visualised through the Cytoscape software. The functional modules were extracted from the PPI network for further analysis. RESULTS: The top 10 hub genes ranked by their connection degrees were IL6, UBE2C, TOP2A, MCM4, PLK2, SMC2, BMP2, LMO7, TRIM36, and MAPK8. Multiple signalling pathways (including the PI3K-Akt signalling pathway, the toll-like receptor signalling pathway, the TNF signalling pathway, and the MAPK pathway) may impact the sensation, transduction, and reaction of external mechanical stimuli. CONCLUSIONS: This study provides a theoretical finding showing that gene UBE2C, IL6, and MAPK8, and multiple signalling pathways may play pivotal roles in dynamic compression-enhanced chondrogenesis.


Assuntos
Condrogênese/genética , Perfilação da Expressão Gênica , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta3/genética , Animais , Bovinos , Expressão Gênica , Interleucina-6 , Transdução de Sinais/genética , Enzimas de Conjugação de Ubiquitina
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(2): 117-122, 2021 Feb 10.
Artigo em Chinês | MEDLINE | ID: mdl-33565061

RESUMO

OBJECTIVE: To compare the mRNA level of cell proliferation-related genes Twist1, SIRT1, FGF2 and TGF-ß3 in placenta mesenchymal stem cells (PA-MSCs), umbilical cord mensenchymals (UC-MSCs) and dental pulp mesenchymal stem cells (DP-MSCs). METHODS: The morphology of various passages of PA-MSCs, UC-MSCs and DP-MSCs were observed by microscopy. Proliferation and promoting ability of the three cell lines were detected with the MTT method. Real-time PCR (RT-PCR) was used to determine the mRNA levels of Twist1, SIRT1, FGF2, TGF-ß3. RESULTS: The morphology of UC-MSCs and DP-MSCs was different from that of PA-MSCs. Proliferation ability and promoting ability of the PA-MSCs was superior to that of UC-MSCs and DP-MSCs. In PA-MSCs, expression level of Twist1 and TGF-ß3 was the highest and FGF2 was the lowest. SIRT1 was highly expressed in UC-MSCs. With the cell subcultured, different expression levels of Twist1, SIRT1, FGF2, TGF-ß3 was observed in PA-MSCs, UC-MSCs and DP-MSCs. CONCLUSION: Up-regulated expression of the Twist1, SIRT1 and TGF-ß3 genes can promote proliferation of PA-MSCs, UC-MSCs and DP-MSCs, whilst TGF-ß3 may inhibit these. The regulatory effect of Twist1, SIRT1, FGF2 and TGF-ß3 genes on PA-MSCs, UC-MSCs and DP-MSCs are different.


Assuntos
Proliferação de Células/genética , Fator 2 de Crescimento de Fibroblastos/genética , Células-Tronco Mesenquimais/citologia , Proteínas Nucleares/genética , Sirtuína 1/genética , Fator de Crescimento Transformador beta3/genética , Proteína 1 Relacionada a Twist/genética , Diferenciação Celular , Células Cultivadas , Polpa Dentária/citologia , Feminino , Humanos , Placenta/citologia , Gravidez , Cordão Umbilical/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA