Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 831
Filter
1.
Front Endocrinol (Lausanne) ; 14: 1167776, 2023.
Article in English | MEDLINE | ID: mdl-37854185

ABSTRACT

Objective: To investigate the clinical and pathological factors associated with preoperative hypothalamus invasion and postoperative outcomes of adamantinomatous craniopharyngiomas (ACPs) after the expanded endonasal approach (EEA) resection. Methods: Ninety-three specimens of ACPs, consisting of 71 primary and 22 recurrent tumors, were investigated for the expression of TGF-ß1, SMAD2, SMAD3, and ß-catenin by immunohistochemistry staining. The clinical information of relevant patients, including the extent of resection, hypothalamus invasion, endocrinopathy, complications, and prognosis, was reviewed. The relationships between the expression of these immunopathological markers and clinical factors were analyzed. Results: Endocrinological dysfunctions were more common in recurrent patients and primary patients with hypothalamus invasion in the comparisons. For recurrent patients, the rate of gross total resection (GTR) was significantly lower than for primary patients (63.6% vs. 90.1%, P = 0.007). According to radiological and intraoperative findings, invasive ACPs (IACPs) included 48 (67.6%) cases in primary tumors. The expression of TGF-ß1 and ß-catenin was significantly higher in recurrent tumors (P = 0.021 and P = 0.018, respectively) and IACPs (P = 0.008 and P = 0.004, respectively). The expression level of TGF-ß1 was associated with hypothalamus involvement (Puget grade, P = 0.05; Vile grade, P = 0.002), postoperative endocrinopathy (P = 0.01), and pituitary stalk preservation (P = 0.008) in primary patients. In addition, the extent of resection, treatment history, hypothalamic invasion, and level of TGF-ß1 expression had significant influences on tumor recurrence/progression after surgery separately. Conclusion: Our study demonstrated the potential role of TGF-ß1 in the regulation of hypothalamus invasion in ACPs and the prediction of prognosis after EEA surgery. The TGF-ß signaling pathway may represent a crucial mechanism in the aggressive behavior and progression of ACPs.


Subject(s)
Craniopharyngioma , Pituitary Neoplasms , Transforming Growth Factor beta1 , Humans , beta Catenin , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Craniopharyngioma/surgery , Neoplasm Recurrence, Local/pathology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Pituitary Neoplasms/surgery , Signal Transduction , Transforming Growth Factor beta1/biosynthesis
2.
Anim Reprod Sci ; 238: 106938, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35124431

ABSTRACT

This study aimed to compare the effect of the administration of either medroxyprogesterone acetate (MPA) or progesterone (P4) in superovulation (SOV) treatments applied during the first follicular wave on follicular development, embryo yield, and the expression of genes related to pluripotency maintenance, differentiation of the trophectoderm, cell growth and differentiation, apoptosis and energy metabolism in sheep embryos. The estrous cycle of 36 multiparous ewes was synchronized with a short protocol, and the animals were randomly allocated to three groups. At the beginning of SOV, 12 ewes per treatment received an intravaginal sponge impregnated with 60 mg of MPA (TMPA), or an intravaginal device containing 0.33 g of P4 (TP4), or received no progestogen treatment (CON). The device was kept until the fifth dose of FSH. Ewes were mated with five fertile rams. Gene expression was performed by RT-qPCR using grade I and II blastocysts. The numbers of corpora lutea, total structures and viable embryos recovered per ewe were similar (P > 0.05) among groups. However, the viability rate was higher in TP4 (71.9 ± 16.3%) compared to CON (24.4 ± 16.8%; P = 0.01) and similar to TMPA (49.9 ± 16.3%; P = 0.2). Similarly, when compared with CON, treatment with P4 or MPA positively regulated the TGFB1 transcript involved in cell proliferation and differentiation (P = 0.01 and P = 0.03, respectively). In conclusion, supplementation with P4 during the first follicular wave of the estrous cycle improves embryo viability and alters the expression of the TGFB1 gene.


Subject(s)
Medroxyprogesterone Acetate , Progesterone , Superovulation , Transforming Growth Factor beta1 , Animals , Dietary Supplements , Embryo, Mammalian , Female , Gene Expression , Male , Medroxyprogesterone Acetate/pharmacology , Ovarian Follicle/drug effects , Pregnancy , Progesterone/pharmacology , Random Allocation , Sheep , Sheep, Domestic , Superovulation/drug effects , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics
3.
Mediators Inflamm ; 2021: 7534601, 2021.
Article in English | MEDLINE | ID: mdl-34373677

ABSTRACT

BACKGROUND: Telocytes (TCs) are a distinct type of interstitial cells that play a vital role in the pathogenesis of ulcerative colitis and colonic tissue hemostasis. The aim of this study was to examine the effect of nanocurcumin (NC) on the morphometric and immunohistochemical characterization of TCs in the ulcerative colitis (UC) rat model. METHODS: Forty rats were randomly divided into control, NC, UC, and UC+NC groups. At the end of the experiment, the colon was dissected and prepared for histopathological and immunohistochemical assessment. Tissue homogenates were prepared for real-time PCR assessment of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta (TGF-ß) gene expression. Our results revealed extensive mucosal damage with inflammatory cell infiltration, significant reduction of CD34, and vimentin immunostained TCs in the colon of the UC group with significant elevation of expression of IL-6, TNF-α, and TGF-ß. The UC+NC-treated group revealed significant elevation of TC count compared to the UC group besides, a significant reduction of the three gene expression. CONCLUSION: NC successfully targeted the colonic tissue, improved the mucosal lesion, preserve TCs distribution, and count through its anti-inflammatory and fibrinolytic properties.


Subject(s)
Colitis, Ulcerative/pathology , Colon/pathology , Curcumin/chemistry , Nanoparticles/chemistry , Telocytes/pathology , Animals , Colitis , Colon/metabolism , Disease Models, Animal , Fibrinolysis , Gene Expression Regulation , Immunohistochemistry , Inflammation , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Male , Rats , Rats, Wistar , Spectroscopy, Fourier Transform Infrared , Transforming Growth Factor beta1/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Vimentin/chemistry
4.
Clin. transl. oncol. (Print) ; 23(7): 1405-1414, jul. 2021. graf
Article in English | IBECS | ID: ibc-221981

ABSTRACT

Purpose Although recent studies have suggested that neutral endopeptidase (NEP) is implicated in the regulation of colon cancer (CC) cell growth and metastasis, the influence of the tumor microenvironment on this role of NEP has not been investigated so far. Normal colon fibroblasts (NCFs) constitute a component of the stroma surrounding a tumor in an early stage of its development. NCFs can influence transformed cells via different paracrine factors, including TGF-β1. This in vitro study was undertaken to evaluate the role of NEP in CC promotion in conditions of indirect co-culture of CC cells (LS180 and SW620) with NCFs (CCD-18Co) or their conditioned medium (CM-18Co). Methods We examined cell proliferation (with the BrdU assay) and invasiveness (using BME-coated inserts, 8 µm) of NEP-expressing, NEP-silenced (siRNA), and NEP-inhibited (with thiorphan, i.e. a NEP specific inhibitor) CC cells cultured alone or co-cultured with CCD-18Co or with their conditioned medium. The Western blot and ELISA methods were used to assess the level of TGF-β1. Results The results showed that the co-culture of the NEP-depleted CC cells with NCFs or their conditioned medium resulted in a significant decrease in cell proliferation in comparison with the proliferative potential of NEP-silenced/inhibited CC cells cultivated alone. In contrast, the NEP depletion did not influence the invasiveness of CC cells in the co-cultures. The co-culture of CC cells with CCD-18Co or CM-C18Co resulted in increased synthesis of TGF-β1, while the NEP downregulation decreased the synthesis of TGF-β1 in CC cells and abolished the stimulatory effect of the co-cultures on TGF-β1 production. Conclusions The results suggest that the expression of NEP by colon cancer cells is essential for their proliferation and TGF-β1 synthesis during paracrine interactions with NCFs (AU)


Subject(s)
Humans , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fibroblasts , Neprilysin/physiology , Transforming Growth Factor beta1/biosynthesis , Tumor Cells, Cultured , Coculture Techniques
5.
Oncol Rep ; 46(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34296292

ABSTRACT

Melanoma is an aggressive type of cancer originating from the skin that arises from neoplastic changes in melanocytes. Transforming growth factor­ß (TGF­ß) is a pleiotropic cytokine and is known to contribute to melanoma progression by inducing the epithelial­mesenchymal transition (EMT) program and creating an environment that favors tumor progression. There are three TGF­ß isoforms, TGF­ß1, TGF­ß2 and TGF­ß3, all of which engage in pro­tumorigenic activities by activating SMAD signaling pathways. All TGF­ß isoforms activate signaling pathways by binding to their TGF­ß type I (TßRI) and type II (TßRII) receptors. Thus, effective targeting of all TGF­ß isoforms is of great importance. In the present study, chimeric proteins comprising the extracellular domains of TßRI and/or TßRII fused with the Fc portion of human immunoglobulin (IgG) were validated in the melanoma context. The Fc chimeric receptor comprising both TßRI and TßRII (TßRI­TßRII­Fc) effectively trapped all TGF­ß isoforms. Conversely, TßRII­Fc chimeric receptor, that comprises TßRII only, was able to interact with TGF­ß1 and TGF­ß3 isoforms, but not with TGF­ß2, which is a poor prognostic factor for melanoma patients. Accordingly, it was revealed that TßRI­TßRII­Fc chimeric receptor suppressed the EMT program in melanoma cells in vitro induced by any of the three TGF­ß isoforms, as revealed by decreased expression of mesenchymal markers. Conversely, TßRII­Fc chimeric receptor inhibited the EMT program induced by TGF­ß1 and TGF­ß3. In addition, it was established that tumor growth in subcutaneous mouse melanoma was inhibited by TßRI­TßRII­Fc chimeric receptor indicating that Fc chimeric receptor could be applied to modify the tumor microenvironment (TME) of melanoma. Therefore, designing of Fc chimeric receptors targeting TGF­ß signals that affect various components of the TME may result in the development of effective anti­melanoma agents.


Subject(s)
Melanoma/metabolism , Receptors, Fc/metabolism , Skin Neoplasms/metabolism , Transforming Growth Factor beta1/biosynthesis , Animals , Cell Proliferation , Cytokines/metabolism , Disease Progression , Epithelial-Mesenchymal Transition , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Melanoma/pathology , Melanoma, Experimental , Mice , Protein Binding , Protein Isoforms , Receptors, Chimeric Antigen/chemistry , Signal Transduction , Skin Neoplasms/pathology , Smad Proteins/metabolism , Tumor Microenvironment
6.
PLoS One ; 16(7): e0253522, 2021.
Article in English | MEDLINE | ID: mdl-34197482

ABSTRACT

BACKGROUND: Glioma is the most common and lethal form of brain cancer. It is highly malignant and is often characterized by chemoresistance and radioresistance, which are thought to mainly result from hypoxic microenvironments. Various tumour-promoting and tumour-suppressing microRNAs (miRNAs) have been identified in gliomas; however, it is still largely unknown how miRNAs are modified by hypoxia and subsequently affect glioma. In this study, we examined the expression of miR-210-3p, a well-characterized miRNA that responds to hypoxia in glioma cell lines. METHODS: The expressions of miR-9 and miR-210-3p were analysed by using qPCR. Cell viability was measured by performing CCK-8 after eechinomycin treatment or introduction of miR-210 for 24 or 48 h. The correlation of HIF-1α expression with TGF-ß were analysed using the REMBRANDT database. The biomarkers of EMT, including E-cadherin, N-cadherin and Vimentin, were detected by western blot. Apoptotic cell death was measured by performing Annexin V-FITC/PI double staining followed by flow cytometry. RESULTS: We found that miR-210-3p was induced by a mechanism dependent on the hypoxia-induced transcriptional activity of HIF-1α. Then we established a positive association between the HIF-1α and TGF-ß expression levels, and miR-210-3p upregulation induced TGF-ß expression, indicating that hypoxia-induced HIF-1α activity upregulated TGF-ß via miR-210-3p upregulation. Hypoxia-induced miR-210-3p activity was found to promote EMT by upregulating TGF-ß, which subsequently enhanced the invasive ability in U87-MG cells. We further confirmed that miR-210-3p induced chemoresistance to TMZ in U87-MG cells via TGF-ß upregulation under hypoxic conditions. CONCLUSION: These results help to reveal the potential regulatory mechanisms of hypoxia-induced miR-210-3p expression that affect malignant behaviors and chemoresistance via TGF-ß upregulation in glioma cells.


Subject(s)
Brain Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Glioma/genetics , MicroRNAs/genetics , Transforming Growth Factor beta1/biosynthesis , Anaerobiosis/physiology , Brain Neoplasms/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Up-Regulation
7.
Biomed Pharmacother ; 141: 111793, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34098216

ABSTRACT

Liposomal honokiol isolated from the genus Magnolia has been found to have antiangiogenic, anti-inflammatory and antitumor properties. However, there has no report on its role in hair growth. Hair follicles are life-long cycled organelles that go through from anagen, catagen and telogen stages and are regulated by diverse signaling pathways, including Wnt/ß-catenin, Notch, Epidermal growth factor (EGF) and Sonic hegehog (SHH). Wnt signals are essential for the initiation of hair follicle placode development and a new potential target of hair loss treatment. This study was designed to investigate the effect of liposomal honokiol (Lip-honokiol) on inducing hair anagen. We identified the hair grew out in advance in the shaving area of C57BL/6N mice after the treatment of liposomal honokiol (Lip-honokiol) by daily abdominal injection. We first demonstrated that Lip-Honokiol activated the Wnt3a/ß-catenin pathway and downregulated the transforming growth factor-ß1 (TGF-ß1) to promote hair growth in mice via immunohistochemistry and immunofluorescence staining. These findings suggest that Lip-honokiol activated the Wnt/ß-catenin pathway and accelerated the transfer from the telogen to anagen stage and finally promoted the hair growth.


Subject(s)
Biphenyl Compounds/administration & dosage , Hair Follicle/drug effects , Hair Follicle/metabolism , Lignans/administration & dosage , Transforming Growth Factor beta1/biosynthesis , Wnt3A Protein/biosynthesis , beta Catenin/biosynthesis , Animals , Down-Regulation/drug effects , Down-Regulation/physiology , Female , Hair/cytology , Hair/drug effects , Hair/metabolism , Hair Follicle/cytology , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta1/antagonists & inhibitors
8.
Int J Biol Sci ; 17(7): 1837-1850, 2021.
Article in English | MEDLINE | ID: mdl-33994866

ABSTRACT

Joint capsule fibrosis caused by excessive inflammation results in post-traumatic joint contracture (PTJC). Transforming growth factor (TGF)-ß1 plays a key role in PTJC by regulating fibroblast functions, however, cytokine-induced TGF-ß1 expression in specific cell types remains poorly characterized. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in inflammation- and fibrosis-associated pathophysiology. In this study, we investigated whether MIF can facilitate TGF-ß1 production from fibroblasts and regulate joint capsule fibrosis following PTJC. Our data demonstrated that MIF and TGF-ß1 significantly increased in fibroblasts of injured rat posterior joint capsules. Treatment the lesion sites with MIF inhibitor 4-Iodo-6-phenylpyrimidine (4-IPP) reduced TGF-ß1 production and relieved joint capsule inflammation and fibrosis. In vitro, MIF facilitated TGF-ß1 expression in primary joint capsule fibroblasts by activating mitogen-activated protein kinase (MAPK) (P38, ERK) signaling through coupling with membrane surface receptor CD74, which in turn affected fibroblast functions and promoted MIF production. Our results reveal a novel function of trauma-induced MIF in the occurrence and development of joint capsule fibrosis. Further investigation of the underlying mechanism may provide potential therapeutic targets for PTJC.


Subject(s)
Intramolecular Oxidoreductases/genetics , Joint Capsule/metabolism , Joint Diseases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/pathology , RNA/genetics , Transforming Growth Factor beta1/genetics , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Intramolecular Oxidoreductases/biosynthesis , Joint Capsule/pathology , Joint Diseases/metabolism , Joint Diseases/pathology , Macrophage Migration-Inhibitory Factors/biosynthesis , Macrophages/metabolism , Male , RNA/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta1/biosynthesis
9.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670400

ABSTRACT

Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer, with only limited treatment options available. Recently, cancer stem cells (CSCs) have emerged as the potential drivers of tumor progression due to their ability to both self-renew and give rise to differentiated progeny. The CSC state has been linked to the process of epithelial-mesenchymal transition (EMT) and to the highly flexible state of epithelial-mesenchymal plasticity (EMP). We aimed to establish primary breast cancer stem cell (BCSC) cultures isolated from TNBC specimens. These cells grow as tumor spheres under anchorage-independent culture conditions in vitro and reliably form tumors in mice when transplanted in limiting dilutions in vivo. The BCSC xenograft tumors phenocopy the original patient tumor in architecture and gene expression. Analysis of an EMT-related marker profile revealed the concomitant expression of epithelial and mesenchymal markers suggesting an EMP state for BCSCs of TNBC. Furthermore, BCSCs were susceptible to stimulation with the EMT inducer TGF-ß1, resulting in upregulation of mesenchymal genes and enhanced migratory abilities. Overall, primary BCSC cultures are a promising model close to the patient that can be used both in vitro and in vivo to address questions of BCSC biology and evaluate new treatment options for TNBC.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , Up-Regulation , Female , Humans , MCF-7 Cells , Neoplasm Proteins/biosynthesis , Neoplastic Stem Cells/pathology , Transforming Growth Factor beta1/biosynthesis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
10.
Clin Transl Oncol ; 23(7): 1405-1414, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33537866

ABSTRACT

PURPOSE: Although recent studies have suggested that neutral endopeptidase (NEP) is implicated in the regulation of colon cancer (CC) cell growth and metastasis, the influence of the tumor microenvironment on this role of NEP has not been investigated so far. Normal colon fibroblasts (NCFs) constitute a component of the stroma surrounding a tumor in an early stage of its development. NCFs can influence transformed cells via different paracrine factors, including TGF-ß1. This in vitro study was undertaken to evaluate the role of NEP in CC promotion in conditions of indirect co-culture of CC cells (LS180 and SW620) with NCFs (CCD-18Co) or their conditioned medium (CM-18Co). METHODS: We examined cell proliferation (with the BrdU assay) and invasiveness (using BME-coated inserts, 8 µm) of NEP-expressing, NEP-silenced (siRNA), and NEP-inhibited (with thiorphan, i.e. a NEP specific inhibitor) CC cells cultured alone or co-cultured with CCD-18Co or with their conditioned medium. The Western blot and ELISA methods were used to assess the level of TGF-ß1. RESULTS: The results showed that the co-culture of the NEP-depleted CC cells with NCFs or their conditioned medium resulted in a significant decrease in cell proliferation in comparison with the proliferative potential of NEP-silenced/inhibited CC cells cultivated alone. In contrast, the NEP depletion did not influence the invasiveness of CC cells in the co-cultures. The co-culture of CC cells with CCD-18Co or CM-C18Co resulted in increased synthesis of TGF-ß1, while the NEP downregulation decreased the synthesis of TGF-ß1 in CC cells and abolished the stimulatory effect of the co-cultures on TGF-ß1 production. CONCLUSIONS: The results suggest that the expression of NEP by colon cancer cells is essential for their proliferation and TGF-ß1 synthesis during paracrine interactions with NCFs.


Subject(s)
Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fibroblasts , Neprilysin/physiology , Transforming Growth Factor beta1/biosynthesis , Cells, Cultured , Coculture Techniques , Humans , Tumor Cells, Cultured
11.
Int J Hematol ; 113(3): 441-455, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33392972

ABSTRACT

Adverse vascular events have become a serious clinical problem in chronic myeloid leukemia (CML) patients who receive certain BCR/ABL1 tyrosine kinase inhibitors (TKIs). Studies have shown that endothelial-to-mesenchymal transition (EndMT) can contribute to various vascular diseases. We investigated the effects of TKIs on the development of EndMT in human vascular-endothelial cells (VECs). Exposure of VECs to dasatinib, but not to other TKIs, produced a significant increase in the formation of spindle-shaped cells. This effect was accompanied by a significant increase in expression of the EndMT inducer transforming growth factor-ß (TGF-ß) and mesenchymal markers vimentin, smooth muscle alpha-actin, and fibronectin, as well as a significant decrease in expression of vascular-endothelial markers CD31 and VE-cadherin attributable at least in part to activation of ERK signaling. Inhibitors of TGF-ß and ERK partially attenuated dasatinib-induced EndMT. Interestingly, bosutinib efficiently counteracted dasatinib-induced EndMT and attenuated dasatinib-induced phosphorylation of ERK. Taken together, these results show that dasatinib induces EndMT, which might contribute to the development of vascular toxicity, such as the pulmonary hypertension observed in CML patients receiving dasatinib. Bosutinib could play a distinct role in protecting VECs from EndMT.


Subject(s)
Aniline Compounds/pharmacology , Cell Transdifferentiation/drug effects , Dasatinib/pharmacology , Endothelial Cells/drug effects , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , A549 Cells , Animals , Biomarkers , Cell Shape/drug effects , Dasatinib/antagonists & inhibitors , Endothelial Cells/cytology , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hybrid Cells , Imatinib Mesylate/pharmacology , Imidazoles/pharmacology , Mesoderm , Mice , Mice, Inbred C57BL , Pyridazines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/genetics , Up-Regulation/drug effects
12.
Eur J Immunol ; 51(1): 125-137, 2021 01.
Article in English | MEDLINE | ID: mdl-33107588

ABSTRACT

Calcitriol and 9-cis retinoic acid (9cRA) play a fundamental role in shaping the adaptive immune response by altering the Ig profile and the differentiation of B cells, controlled by their corresponding nuclear receptors, VDR and RAR. Herein, after the establishment of a plasmablast differentiation culture, we investigated how both ligands modulate human naïve B cell differentiation and to which extent VDR/RXR and RAR/RXR signaling interferes. Calcitriol and 9cRA mediated activation of purified naïve B cells resulted in a strong differentiation of CD27+ CD38+ plasmablasts and antibody secretion. The significant IgA response was preceded by a strong induction of α-germline transcription (GLT). Induction of αGLT and consecutively IgA secretion driven by calcitriol is a novel observation and we show by magnetic chromatin IP that this was mediated by recruitment of the VDR to the TGF-ß promoter thus inducing TGF-ß expression. Finally, as revealed by transcriptomic profiling calcitriol and 9cRA modulate several signals required for differentiation and isotype switching in a noncompeting but rather additive manner. Calcitriol and 9cRA participate in the control of the IgA response in human activated naïve B cells. The balance between both ligands may be an important factor in channeling humoral immune responses toward a protective direction.


Subject(s)
Alitretinoin/immunology , Alitretinoin/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Calcitriol/immunology , Calcitriol/pharmacology , Immunoglobulin A/biosynthesis , Adaptive Immunity/drug effects , B-Lymphocytes/cytology , Binding Sites/genetics , CD40 Ligand/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , GTP-Binding Proteins/genetics , Gene Expression , Humans , Immunoglobulin Class Switching/drug effects , Immunoglobulin Class Switching/immunology , Interleukin-4/immunology , Ligands , Lymphocyte Activation , Plasma Cells/cytology , Plasma Cells/drug effects , Plasma Cells/immunology , Promoter Regions, Genetic , Protein Glutamine gamma Glutamyltransferase 2 , Receptors, Calcitriol/immunology , Receptors, Retinoic Acid/immunology , Retinoid X Receptors/immunology , Signal Transduction/immunology , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics , Transglutaminases/genetics , Vitamin D3 24-Hydroxylase/genetics
13.
Sci Rep ; 10(1): 19089, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154445

ABSTRACT

Exaggerated transforming growth factor-beta 1 (TGFß1) expression worsens fibroproliferation following bleomycin-induced lung injury in alcohol-fed mice. MicroRNA (miR)-1946a is predicted to bind to the TGFß1 3' untranslated region (UTR), thereby inhibiting its transcription. We hypothesize that alcohol suppresses miR-1946a and induces TGFß1. Primary murine lung fibroblasts (PLFs) were cultured ± alcohol, miR-1946a mimic or inhibitor, and TGFß1 signaling inhibitors. miR-1946a was analyzed after alcohol treatment in vitro and in vivo. TGFß1 expression and TGFß1 3'UTR-luciferase activity was quantified. We showed that alcohol suppressed miR-1946a in the alcohol-fed mouse lungs and PLFs. MiR-1946a inhibitor increased TGFß1 expression in the fibroblast. MiR-1946a mimic treatment suppressed TGFß1 gene expression and TGFß1 3'UTR activity. Overexpression of miR1946a inhibited alcohol-induced TGFß1 gene and protein expression as well as alcohol-induced TGFß1 and α-smooth muscle actin (SMA) protein expression in PLFs. In conclusion, miR-1946a modulates TGFß1 expression through direct interaction with TGFß1 3'UTR. These findings identify a novel mechanism by which alcohol induces TGFß1 in the lung.


Subject(s)
Ethanol/toxicity , Lung/drug effects , Lung/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics , 3' Untranslated Regions , Actins/genetics , Actins/metabolism , Alcoholism/genetics , Alcoholism/metabolism , Alcoholism/pathology , Animals , Cells, Cultured , Down-Regulation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Lung/pathology , Lung Injury/chemically induced , Lung Injury/genetics , Lung Injury/metabolism , Mice , Mice, Inbred C57BL
14.
Drug Des Devel Ther ; 14: 4315-4326, 2020.
Article in English | MEDLINE | ID: mdl-33116418

ABSTRACT

BACKGROUND: N-Acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a short peptide with an anti-silicosis effect. However, the short biological half-life and low plasma concentration of Ac-SDKP hamper discovery of specific targets in organisms and reduce the anti-silicosis effect. A novel peptide, Ac-SDK (biotin) proline, termed "Ac-B", with anti-fibrotic properties was synthesized. METHODS: Ac-B was detected quantitatively by high-performance liquid chromatography. Phagocytosis of Ac-B by the alveolar epithelial cell line A549 was investigated by confocal laser scanning microscopy and flow cytometry. To further elucidate the cellular-uptake mechanism of Ac-B, chemical inhibitors of specific uptake pathways were used. After stimulation with transforming growth factor-ß1, the effects of Ac-B on expression of the myofibroblast marker vimentin and accumulation of collagen type I in A549 cells were analyzed by Western blotting. Sirius Red staining and immunohistochemical analyses of the effect of Ac-B on expression of α-smooth muscle actin (SMA) in a rat model of silicosis were undertaken. RESULTS: Ac-B had good traceability during the uptake, entry, and distribution in cells. Ac-B treatment prevented an increase in α-SMA expression in vivo and in vitro and was superior to that of Ac-SDKP. Caveolae-mediated uptake of Ac-B by A549 cells led to achieving anti-epithelial-mesenchymal transformation (EMT) effects. CONCLUSION: Ac-B had an anti-fibrotic effect and could be a promising agent for the fibrosis observed in silicosis in the future.


Subject(s)
Oligopeptides/chemistry , Pulmonary Fibrosis/drug therapy , Silicosis/drug therapy , A549 Cells , Actins/metabolism , Animals , Chromatography, High Pressure Liquid , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung/pathology , Male , Oligopeptides/pharmacokinetics , Phagocytosis , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , Rats , Rats, Wistar , Silicosis/complications , Silicosis/pathology , Transforming Growth Factor beta1/biosynthesis
15.
Cell Death Dis ; 11(9): 765, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943605

ABSTRACT

TGFß1 signaling is a critical driver of collagen accumulation in pulmonary fibrotic diseases and a well-characterized regulator of cancer associated fibroblasts (CAF) activation in lung cancer. Myofibroblasts induced by TGFß1 and other factors are key players in the pathogenesis of lung fibrosis and tumor. Tremendous attention has been gained to targeting myofibroblasts in order to inhibit the progression of fibrosis and myofibroblast-induced tumor progression and metastasis. Here we determined the therapeutic efficacy of simultaneously targeting PI3K and HDAC pathways in lung myofibroblasts and CAF with a single agent and to evaluate biomarkers of treatment response. CUDC-907 is a first-in-class compound, functioning as a dual inhibitor of HDACs and PI3K/AKT pathway. We investigated its effects in counteracting the activity of TGFß1-induced myofibroblasts/CAF in regard to cell proliferation, migration, invasion, apoptosis in vitro antifibrosis efficiency in vivo. We found that CUDC-907 inhibited myofibroblasts/CAF cell proliferation, migration and apoptosis in a dose-dependent manner and caused cell cycle arrest at G1-S phase. CUDC-907 not only inhibited myofibroblasts markers expression, but also significantly inhibited the phosphorylation level of AKT, mTOR, Smad2/3, and promoted acetylation of histones. Furthermore, the observed inhibitory effect was also confirmed in bleomycin-induced mice lung fibrosis and nude mouse transplanted tumor model. Overall, these data suggest that dual inhibition of HDAC and the tyrosine kinase signaling pathways with CUDC-907 is a promising treatment strategy for TGFß1-induced lung and tumor fibrosis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Fibrosis/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung/metabolism , Morpholines/pharmacology , Pyrimidines/pharmacology , Transforming Growth Factor beta1/biosynthesis , Aged , Animals , Apoptosis , Bleomycin/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Female , Fibroblasts/metabolism , Fibrosis/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , NIH 3T3 Cells , Neoplasm Transplantation , Protein-Tyrosine Kinases/antagonists & inhibitors
16.
J Reprod Dev ; 66(6): 493-504, 2020 Dec 22.
Article in English | MEDLINE | ID: mdl-32801258

ABSTRACT

Circular RNA (circRNA) plays a key role in the development and progression of several diseases; however, its role in intrauterine adhesions (IUAs) is not well understood. This study aims to investigate the expression profiles and potential role of circRNA in IUA. RNA-sequencing was performed to screen for abnormally expressed circRNAs in TGF-ß1-induced IUA endometrial stromal cell (ESC) model (IUA group) and an SMAD3 inhibitor, SIS3-treated IUA ESC model (SIS3 group). Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed to uncover the key functions and pathways. Interaction networks were constructed and analyzed based on the competing endogenous RNA hypothesis of circRNA. CircRNAs were validated by Sanger sequencing and quantitative polymerase chain reaction (qPCR). Cell proliferation and apoptosis were measured using MTS and flow cytometry, respectively. The protein and mRNA expression levels of fibrosis-related proteins were measured using western blotting and reverse transcription-qPCR, respectively. A total of 66 circRNAs were differentially expressed between the IUA and SIS3 groups. CircPlekha7 was identified as one of the significantly upregulated circRNAs in the SIS3 group. Overexpression of circPlekha7 enhanced apoptosis, decreased the viability of ESCs, and suppressed the expression of α-SMA, collagen I, and SMAD3 in ESCs; whereas knockdown of circPlekha7 exhibited opposite results. Altogether, the results indicate that circPlekha7 plays an anti-fibrotic role in IUA and may serve as a promising prognostic biomarker for patients with IUA. Therefore, overexpression of circPlekha7 could be a potential treatment strategy for IUA.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Endometrium/metabolism , RNA, Circular , Stromal Cells/metabolism , Tissue Adhesions/metabolism , Animals , Carrier Proteins/genetics , Cell Proliferation , Computational Biology , Epithelial Cells/metabolism , Female , Fibrosis , Gene Expression Profiling , Gene Expression Regulation , RNA-Seq , Rats , Smad3 Protein/antagonists & inhibitors , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/metabolism
17.
J Clin Invest ; 130(8): 4360-4369, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32352930

ABSTRACT

Retinitis pigmentosa (RP) is a genetically heterogenous group of eye diseases in which initial degeneration of rods triggers secondary degeneration of cones, leading to significant loss of daylight, color, and high-acuity vision. Gene complementation with adeno-associated viral (AAV) vectors is one strategy to treat RP. Its implementation faces substantial challenges, however; for example, the tremendous number of loci with causal mutations. Gene therapy targeting secondary cone degeneration is an alternative approach that could provide a much-needed generic treatment for many patients with RP. Here, we show that microglia are required for the upregulation of potentially neurotoxic inflammatory factors during cone degeneration in RP, creating conditions that might contribute to cone dysfunction and death. To ameliorate the effects of such factors, we used AAV vectors to express isoforms of the antiinflammatory cytokine transforming growth factor beta (TGF-ß). AAV-mediated delivery of TGF-ß1 rescued degenerating cones in 3 mouse models of RP carrying different pathogenic mutations. Treatment with TGF-ß1 protected vision, as measured by 2 behavioral assays, and could be pharmacologically disrupted by either depleting microglia or blocking the TGF-ß receptors. Our results suggest that TGF-ß1 may be broadly beneficial for patients with cone degeneration, and potentially other forms of neurodegeneration, through a pathway dependent upon microglia.


Subject(s)
Microglia/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinitis Pigmentosa , Transforming Growth Factor beta1/biosynthesis , Animals , Dependovirus , Disease Models, Animal , Genetic Vectors , Humans , Mice , Mice, Transgenic , Microglia/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/prevention & control , Transduction, Genetic , Transforming Growth Factor beta1/genetics
18.
Brain Res Bull ; 161: 21-32, 2020 08.
Article in English | MEDLINE | ID: mdl-32389801

ABSTRACT

Traumatic brain injury (TBI) causes substantial mortality and long-term disability worldwide. TGFß1 is a unique molecular and functional signature in microglia, but the role of TGFß1 in TBI is not clear. The purpose of this study was to investigate the role of TGFß1 in TBI. The weight dropping device was used to establish TBI model of rats. Hematoxylin eosin staining and Bielschowsky silver staining were used to assess tissue loss. Beam walking and muscle strength tests were used to assess neurological deficits. Immunohistochemical staining was used to assess axonal injures. Western blotting was used to detect expression of related proteins. RT-PCR was used to detect expression of cytokines. Immunofluorescence staining was used to assess the microglia/macrophages activation. We observed obvious axonal injury and microglia/macrophages activation in the peri-lesion cortex. The expression of inflammatory cytokines was markedly high after TBI. The expression of TGFß1 and TGFßRI were significantly reduced after TBI. TGFß1 promoted the functional recovery and alleviated axonal injury 1 day after TBI. TGFß1 promoted microglia/macrophages polarizing to alternative activation and alleviated neuroinflammation. These effects of TGFß1 could be inhibited by LY2109761, the inhibitor of TGFRI/II. These results suggested that TGFß1 played a protective role in axonal injury and could be a potential therapeutic target in early stages following TBI.


Subject(s)
Axons/metabolism , Brain Injuries, Traumatic/metabolism , Macrophages/metabolism , Microglia/metabolism , Transforming Growth Factor beta1/biosynthesis , Animals , Axons/drug effects , Axons/pathology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/prevention & control , Macrophages/drug effects , Macrophages/pathology , Male , Microglia/drug effects , Microglia/pathology , Pyrazoles/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley
19.
Biomed Pharmacother ; 127: 110119, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32276127

ABSTRACT

Idiopathic pulmonary fibrosis (IPF), characterized by excessive collagen deposition, is a progressive and typically fatal lung disease without effective therapeutic strategies. Juglanin, as a natural product mainly isolated from green walnut husks of Juglans mandshuric, has various bioactivities, including anti-oxidative, anti-inflammatory and anti-fibrotic effects. Stimulator of interferon genes (Sting) is a signaling molecule and plays an essential role in meditating fibrosis. However, the effects of Jug and Sting on pulmonary fibrosis are not fully understood. In this study, we investigated the role of Jug in bleomycin (BLM)-induced inflammation and fibrosis mouse model, as well as the underlying molecular mechanism. The results here indicated that Jug-treated mice exhibited a definitively improved survival rate than that of the BLM-challenged mice. Jug administration significantly alleviated neutrophil alveolar infiltration, lung vascular permeability and pro-inflammatory response in BLM mice. Subsequently, the pulmonary fibrosis induced by BLM was markedly attenuated by Jug through reducing the expression of fibrotic hallmarks, including transforming growth factor-ß1 (TGF-ß1), fibronectin, matrix metallo-proteinase-9 (MMP-9), α-smooth muscle actin (α-SMA) and collagen I. Importantly, we found that BLM mice showed higher expression levels of Sting in lung tissues, which were notably restrained by Jug treatment. The role of Jug in suppressing Sting was confirmed in TGF-ß-incubated cells. Notably, the in vitro analysis further showed that Sting knockdown could ameliorate TGF-ß-triggered collagen accumulation. In contrast, TGF-ß-induced fibrosis was accelerated by Sting over-expression. Therefore, BLM may induce lung fibrosis through activating Sting signaling, and Jug could be used therapeutically to improve tissue repair and attenuate the intractable disease.


Subject(s)
Glycosides/pharmacology , Inflammation/prevention & control , Kaempferols/pharmacology , Membrane Proteins/metabolism , Pulmonary Fibrosis/prevention & control , Actins/biosynthesis , Animals , Bleomycin , Collagen Type I/biosynthesis , Fibronectins/biosynthesis , Gene Knockdown Techniques , Inflammation/complications , Male , Matrix Metalloproteinase 9/biosynthesis , Membrane Proteins/genetics , Mice , Pulmonary Fibrosis/chemically induced , Signal Transduction/drug effects , Survival Rate , Transforming Growth Factor beta1/biosynthesis
20.
Neurochirurgie ; 66(3): 168-173, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32201238

ABSTRACT

PURPOSE: An accurate understanding of cellular biochemical changes in human intervertebral disc (IVD)s and the corresponding mechanisms during the developmental process still remain unknown and important for investigating the function of critical factors in normal IVD development as well as ascertaining the therapeutic targets for the IVD degeneration. METHODS: Under ethical conditions, human fetal cervical IVDs at 4, 5, and 6 months of pregnancy were collected at abortion surgery. Normal adult human C3-C7 cervical IVDs were taken from cadaveric donors. Sox9, Pax1, TGF-ß1 and type I/II collagen protein and RNA were detected. The number of positive cells was counted to calculate the optical density value for each factor. RESULTS: Sox9, Pax1, and TGF-ß1 expression in the IVD was remarkably reduced with the developmental stage. The location of high expression of Sox9, Pax1, and TGF-ß1 changed with the developmental stage, and migrated from the nucleus pulposus to the annulus fibrosus and endplate. Higher Sox9, Pax1, and TGF-ß1 expression was finally observed around the sclerotome of the vertebral body. The anabolism of type I/II collagens is significantly increased in the IVD in the mid-trimester fetus. CONCLUSIONS: Sox9, Pax1 and TGF-ß1 participate in the developmental process of the human IVD and vertebral body. However, these factors show a separate expression of mRNA and protein, suggesting that they are expressed in the strict time and spatial order.


Subject(s)
Collagen Type II/biosynthesis , Collagen Type I/biosynthesis , Intervertebral Disc/growth & development , Intervertebral Disc/metabolism , Paired Box Transcription Factors/biosynthesis , SOX9 Transcription Factor/biosynthesis , Transforming Growth Factor beta1/biosynthesis , Adult , Cadaver , Collagen Type I/genetics , Collagen Type II/genetics , Female , Humans , Immunohistochemistry , Intervertebral Disc/embryology , Intervertebral Disc Degeneration , Paired Box Transcription Factors/genetics , Pregnancy , Pregnancy Trimester, Second , RNA/biosynthesis , RNA/genetics , SOX9 Transcription Factor/genetics , Transforming Growth Factor beta1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL