Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 5.882
1.
Adv Exp Med Biol ; 1408: 309-328, 2023.
Article En | MEDLINE | ID: mdl-37093435

Transforming growth factor-beta1 (TGF-ß) regulates a plethora of cell-intrinsic processes that modulate tumor progression in a context-dependent manner. Thus, although TGF-ß acts as a tumor suppressor in the early stages of tumorigenesis, in late stages, this factor promotes tumor progression and metastasis. In addition, TGF-ß also impinges on the tumor microenvironment by modulating the immune system. In this aspect, TGF-ß exhibits a potent immunosuppressive effect, which allows both cancer cells to escape from immune surveillance and confers resistance to immunotherapy. While TGF-ß inhibits the activation and antitumoral functions of T-cell lymphocytes, dendritic cells, and natural killer cells, it promotes the generation of T-regulatory cells and myeloid-derived suppressor cells, which hinder antitumoral T-cell activities. Moreover, TGF-ß promotes tumor-associated macrophages and neutrophils polarization from M1 into M2 and N1 to N2, respectively. Altogether, these effects contribute to the generation of an immunosuppressive tumor microenvironment and support tumor promotion. This review aims to analyze the relevant evidence on the complex role of TGF-ß in cancer immunology, the current outcomes of combined immunotherapies, and the anti-TGF-ß therapies that may improve the success of current and new oncotherapies.


T-Lymphocytes, Regulatory , Transforming Growth Factor beta1 , Humans , Killer Cells, Natural , Carcinogenesis , Immunotherapy , Transforming Growth Factor beta/physiology , Tumor Microenvironment
2.
Annu Rev Physiol ; 85: 269-291, 2023 02 10.
Article En | MEDLINE | ID: mdl-36266260

Myostatin (GDF-8) was discovered 25 years ago as a new transforming growth factor-ß family member that acts as a master regulator of skeletal muscle mass. Myostatin is made by skeletal myofibers, circulates in the blood, and acts back on myofibers to limit growth. Myostatin appears to have all of the salient properties of a chalone, which is a term proposed over a half century ago to describe hypothetical circulating, tissue-specific growth inhibitors that control tissue size. The elucidation of the molecular, cellular, and physiological mechanisms underlying myostatin activity suggests that myostatin functions as a negative feedback regulator of muscle mass and raises the question as to whether this type of chalone mechanism is unique to skeletal muscle or whether it also operates in other tissues.


Chalones , Myostatin , Humans , Muscle, Skeletal/physiology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology
3.
Nat Rev Nephrol ; 18(9): 545-557, 2022 09.
Article En | MEDLINE | ID: mdl-35788561

Kidney fibrosis, characterized by excessive deposition of extracellular matrix (ECM) that leads to tissue scarring, is the final common outcome of a wide variety of chronic kidney diseases. Rather than being distributed uniformly across the kidney parenchyma, renal fibrotic lesions initiate at certain focal sites in which the fibrogenic niche is formed in a spatially confined fashion. This niche provides a unique tissue microenvironment that is orchestrated by a specialized ECM network consisting of de novo-induced matricellular proteins. Other structural elements of the fibrogenic niche include kidney resident and infiltrated inflammatory cells, extracellular vesicles, soluble factors and metabolites. ECM proteins in the fibrogenic niche recruit soluble factors including WNTs and transforming growth factor-ß from the extracellular milieu, creating a distinctive profibrotic microenvironment. Studies using decellularized ECM scaffolds from fibrotic kidneys show that the fibrogenic niche autonomously promotes fibroblast proliferation, tubular injury, macrophage activation and endothelial cell depletion, pathological features that recapitulate key events in the pathogenesis of chronic kidney disease. The concept of the fibrogenic niche represents a paradigm shift in understanding of the mechanism of kidney fibrosis that could lead to the development of non-invasive biomarkers and novel therapies not only for chronic kidney disease, but also for fibrotic diseases of other organs.


Kidney , Renal Insufficiency, Chronic , Extracellular Matrix , Fibrosis , Humans , Kidney/pathology , Renal Insufficiency, Chronic/metabolism , Transforming Growth Factor beta/physiology
5.
Sci Rep ; 12(1): 3080, 2022 02 23.
Article En | MEDLINE | ID: mdl-35197532

Fibrosis is a leading cause of morbidity and mortality worldwide. Although fibrosis may involve different organ systems, transforming growth factor-ß (TGFß) has been established as a master regulator of fibrosis across organs. Pirfenidone and Nintedanib are the only currently-approved drugs to treat fibrosis, specifically idiopathic pulmonary fibrosis, but their mechanisms of action remain poorly understood. To identify novel drug targets and uncover potential mechanisms by which these drugs attenuate fibrosis, we performed an integrative 'omics analysis of transcriptomic and proteomic responses to TGFß1-stimulated lung fibroblasts. Significant findings were annotated as associated with pirfenidone and nintedanib treatment in silico via Coremine. Integrative 'omics identified a co-expressed transcriptomic and proteomic module significantly correlated with TGFß1 treatment that was enriched (FDR-p = 0.04) with genes associated with pirfenidone and nintedanib treatment. While a subset of genes in this module have been implicated in fibrogenesis, several novel TGFß1 signaling targets were identified. Specifically, four genes (BASP1, HSD17B6, CDH11, and TNS1) have been associated with pirfenidone, while five genes (CLINT1, CADM1, MTDH, SYDE1, and MCTS1) have been associated with nintedanib, and MYDGF has been implicated with treatment using both drugs. Using the Clue Drug Repurposing Hub, succinic acid was highlighted as a metabolite regulated by the protein encoded by HSD17B6. This study provides new insights into the anti-fibrotic actions of pirfenidone and nintedanib and identifies novel targets for future mechanistic studies.


Antifibrotic Agents/pharmacology , Computational Biology/methods , Extracellular Matrix Proteins/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Indoles/pharmacology , Pyridones/pharmacology , Transforming Growth Factor beta/physiology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Antifibrotic Agents/therapeutic use , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion Molecule-1/genetics , Cell Adhesion Molecule-1/metabolism , Female , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/therapeutic use , Interleukins/genetics , Interleukins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pyridones/therapeutic use , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tensins/genetics , Tensins/metabolism
6.
Zhonghua Nan Ke Xue ; 28(9): 843-847, 2022 Sep.
Article Zh | MEDLINE | ID: mdl-37839012

Benign prostatic hyperplasia (BPH) is a most common disorder in the elderly. The development and growth of the prostate are regulated by androgens and growth factors, and the latter are secreted by prostatic epithelial and mesenchymal cells and play a key role. As the family members of FGF and TGF-ß, FGF2 and TGF-ß1 are the most representative growth factors in the prostate, involved in the regulation of such biological processes as proliferation, apoptosis, migration and differentiation of prostate cells, which provides a theoretical basis for the clinical application of FGF- and TGF-ß-targeted drugs in the treatment of BPH.


Prostatic Hyperplasia , Transforming Growth Factor beta1 , Male , Humans , Aged , Transforming Growth Factor beta1/metabolism , Prostatic Hyperplasia/metabolism , Fibroblast Growth Factor 2/metabolism , Prostate/metabolism , Transforming Growth Factor beta/physiology
7.
Mol Psychiatry ; 27(1): 296-306, 2022 01.
Article En | MEDLINE | ID: mdl-34131268

Neuropsychiatric diseases are manifested by maladaptive behavioral plasticity. Despite the greater understanding of the neuroplasticity underlying behavioral adaptations, pinpointing precise cellular mediators has remained elusive. This has stymied the development of pharmacological interventions to combat these disorders both at the level of progression and relapse. With increased knowledge on the putative role of the transforming growth factor (TGF- ß) family of proteins in mediating diverse neuroadaptations, the influence of TGF-ß signaling in regulating maladaptive cellular and behavioral plasticity underlying neuropsychiatric disorders is being increasingly elucidated. The current review is focused on what is currently known about the TGF-ß signaling in the central nervous system in mediating cellular and behavioral plasticity related to neuropsychiatric manifestations.


Mental Disorders , Nervous System Diseases , Signal Transduction , Transforming Growth Factor beta , Central Nervous System , Humans , Neuronal Plasticity , Transforming Growth Factor beta/physiology
8.
Front Immunol ; 12: 774807, 2021.
Article En | MEDLINE | ID: mdl-34925345

Radiation-induced lung injury (RILI) is a form of radiation damage to normal lung tissue caused by radiotherapy (RT) for thoracic cancers, which is most commonly comprised of radiation pneumonitis (RP) and radiation pulmonary fibrosis (RPF). Moreover, with the widespread utilization of immunotherapies such as immune checkpoint inhibitors as first- and second-line treatments for various cancers, the incidence of immunotherapy-related lung injury (IRLI), a severe immune-related adverse event (irAE), has rapidly increased. To date, we know relatively little about the underlying mechanisms and signaling pathways of these complications. A better understanding of the signaling pathways may facilitate the prevention of lung injury and exploration of potential therapeutic targets. Therefore, this review provides an overview of the signaling pathways of RILI and IRLI and focuses on their crosstalk in diverse signaling pathways as well as on possible mechanisms of adverse events resulting from combined radiotherapy and immunotherapy. Furthermore, this review proposes potential therapeutic targets and avenues of further research based on signaling pathways. Many new studies on pyroptosis have renewed appreciation for the value and importance of pyroptosis in lung injury. Therefore, the authors posit that pyroptosis may be the common downstream pathway of RILI and IRLI; discussion is also conducted regarding further perspectives on pyroptosis as a crucial signaling pathway in lung injury treatment.


Immune Checkpoint Inhibitors/adverse effects , Lung Injury/etiology , Pulmonary Fibrosis/etiology , Radiation Pneumonitis/etiology , HMGB1 Protein/physiology , Humans , NF-E2-Related Factor 2/physiology , Pyroptosis , Signal Transduction/physiology , Transforming Growth Factor beta/physiology
9.
Front Immunol ; 12: 768771, 2021.
Article En | MEDLINE | ID: mdl-34790203

Osteoarticular diseases (OD), such as rheumatoid arthritis (RA) and osteoarthritis (OA) are chronic autoimmune/inflammatory and age-related diseases that affect the joints and other organs for which the current therapies are not effective. Cell therapy using mesenchymal stem/stromal cells (MSCs) is an alternative treatment due to their immunomodulatory and tissue differentiation capacity. Several experimental studies in numerous diseases have demonstrated the MSCs' therapeutic effects. However, MSCs have shown heterogeneity, instability of stemness and differentiation capacities, limited homing ability, and various adverse responses such as abnormal differentiation and tumor formation. Recently, acellular therapy based on MSC secreted factors has raised the attention of several studies. It has been shown that molecules embedded in extracellular vesicles (EVs) derived from MSCs, particularly those from the small fraction enriched in exosomes (sEVs), effectively mimic their impact in target cells. The biological effects of sEVs critically depend on their cargo, where sEVs-embedded microRNAs (miRNAs) are particularly relevant due to their crucial role in gene expression regulation. Therefore, in this review, we will focus on the effect of sEVs derived from MSCs and their miRNA cargo on target cells associated with the pathology of RA and OA and their potential therapeutic impact.


Arthritis, Rheumatoid/therapy , Extracellular Vesicles/physiology , Mesenchymal Stem Cell Transplantation , MicroRNAs/physiology , Osteoarthritis/therapy , Arthritis, Rheumatoid/etiology , Humans , Osteoarthritis/etiology , Transforming Growth Factor beta/physiology
10.
J Clin Invest ; 131(22)2021 11 15.
Article En | MEDLINE | ID: mdl-34779414

Cerebral small vessel disease (CSVD) causes dementia and gait disturbance due to arteriopathy. Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is a hereditary form of CSVD caused by loss of high-temperature requirement A1 (HTRA1) serine protease activity. In CARASIL, arteriopathy causes intimal thickening, smooth muscle cell (SMC) degeneration, elastic lamina splitting, and vasodilation. The molecular mechanisms were proposed to involve the accumulation of matrisome proteins as substrates or abnormalities in transforming growth factor ß (TGF-ß) signaling. Here, we show that HTRA1-/- mice exhibited features of CARASIL-associated arteriopathy: intimal thickening, abnormal elastic lamina, and vasodilation. In addition, the mice exhibited reduced distensibility of the cerebral arteries and blood flow in the cerebral cortex. In the thickened intima, matrisome proteins, including the hub protein fibronectin (FN) and latent TGF-ß binding protein 4 (LTBP-4), which are substrates of HTRA1, accumulated. Candesartan treatment alleviated matrisome protein accumulation and normalized the vascular distensibility and cerebral blood flow. Furthermore, candesartan reduced the mRNA expression of Fn1, Ltbp-4, and Adamtsl2, which are involved in forming the extracellular matrix network. Our results indicate that these accumulated matrisome proteins may be potential therapeutic targets for arteriopathy in CARASIL.


Alopecia/drug therapy , Benzimidazoles/therapeutic use , Biphenyl Compounds/therapeutic use , Cerebral Infarction/drug therapy , High-Temperature Requirement A Serine Peptidase 1/physiology , Leukoencephalopathies/drug therapy , Spinal Diseases/drug therapy , Tetrazoles/therapeutic use , ADAMTS Proteins/analysis , Alopecia/complications , Animals , Cerebral Infarction/complications , Cerebrovascular Circulation/drug effects , Disease Progression , Extracellular Matrix Proteins/analysis , Latent TGF-beta Binding Proteins/analysis , Leukoencephalopathies/complications , Mice , Mice, Inbred C57BL , Recombinant Proteins/analysis , Spinal Diseases/complications , Transforming Growth Factor beta/physiology
11.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article En | MEDLINE | ID: mdl-34681813

Idiopathic pulmonary fibrosis (IPF) is characterized by fibrotic change in alveolar epithelial cells and leads to the irreversible deterioration of pulmonary function. Transforming growth factor-beta 1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) in type 2 lung epithelial cells contributes to excessive collagen deposition and plays an important role in IPF. Atractylodin (ATL) is a kind of herbal medicine that has been proven to protect intestinal inflammation and attenuate acute lung injury. Our study aimed to determine whether EMT played a crucial role in the pathogenesis of pulmonary fibrosis and whether EMT can be utilized as a therapeutic target by ATL treatment to mitigate IPF. To address this topic, we took two steps to investigate: 1. Utilization of anin vitro EMT model by treating alveolar epithelial cells (A549 cells) with TGF-ß1 followed by ATL treatment for elucidating the underlying pathways, including Smad2/3 hyperphosphorylation, mitogen-activated protein kinase (MAPK) pathway overexpression, Snail and Slug upregulation, and loss of E-cadherin. Utilization of an in vivo lung injury model by treating bleomycin on mice followed by ATL treatment to demonstrate the therapeutic effectiveness, such as, less collagen deposition and lower E-cadherin expression. In conclusion, ATL attenuates TGF-ß1-induced EMT in A549 cells and bleomycin-induced pulmonary fibrosis in mice.


Alveolar Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Furans/pharmacology , Idiopathic Pulmonary Fibrosis/prevention & control , A549 Cells , Alveolar Epithelial Cells/physiology , Animals , Bleomycin/adverse effects , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Furans/therapeutic use , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
12.
Int Heart J ; 62(5): 1135-1144, 2021.
Article En | MEDLINE | ID: mdl-34588407

Myocardial fibrosis is an important pathological phenomenon of cardiac remodeling that is induced by hypertension, myocardial ischemia, valvular heart disease, hypertrophic cardiomyopathy, and other heart diseases and can progress to heart failure. Urotensin II (UII) is regarded as a cardiovascular autacoid/hormone that is not only the most potent vasoconstrictor in mammals but also involved in cardiac remodeling. However, the molecular mechanisms responsible for UII-induced cardiac fibrosis have not yet been fully elucidated. Therefore, we aimed to investigate the effect of UII on myocardial fibrosis in cardiac hypertrophy and the mechanism of UII-induced cardiac fibrosis. Cardiac tissue from mice subjected to Transverse aortic constriction (TAC) was collected. Cardiac hypertrophy, myocardial fibrosis, and the expression of UII protein were assessed using echocardiography and pathological and molecular biological analyses. The effect of UII on fibrosis was evaluated in UII-treated mice and isolated rat primary cardiac fibroblasts, and the results indicated that UII induced significant myocardial fibrosis and increases in the proliferation and fibrotic responses both in mice and cultured fibroblasts. Mechanistically, UII treatment induced activation of the TGF-ß/Smad signaling pathway, which was suppressed by the UII receptor antagonist. In conclusion, UII plays critical roles in cardiac fibrosis by modulating the TGF-ß/Smads signaling pathway, which may be a promising therapeutic target in hypertrophic cardiomyopathy and related problems, such as cardiac remodeling and heart failure.


Cardiomegaly/etiology , Myocardium/pathology , Signal Transduction , Smad1 Protein/physiology , Transforming Growth Factor beta/physiology , Urotensins/adverse effects , Animals , Fibrosis/chemically induced , Male , Mice , Mice, Inbred C57BL
13.
Front Immunol ; 12: 708874, 2021.
Article En | MEDLINE | ID: mdl-34484208

Regulatory T cells (Treg) are essential to maintain immune homeostasis and prevent autoimmune disorders. While the function and molecular regulation of Foxp3+CD4+ Tregs are well established, much of CD8+ Treg biology remains to be revealed. Here, we will review the heterogenous subsets of CD8+ T cells have been named "CD8+ Treg" and mainly focus on CD122hiLy49+CD8+ Tregs present in naïve mice. CD122hiLy49+CD8+ Tregs, which depends on transcription factor Helios and homeostatic cytokine IL-15, have been established as a non-redundant regulator of germinal center (GC) reaction. Recently, we have demonstrated that TGF-ß (Transforming growth factor-ß) and transcription factor Eomes (Eomesodermin) are essential for the function and homeostasis of CD8+ Tregs. In addition, we will discuss several open questions regarding the differentiation, function and true identity of CD8+ Tregs as well as a brief comparison between two regulatory T cell subsets critical to control GC reaction, namely CD4+ TFR (follicular regulatory T cells) and CD8+ Tregs.


CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/classification , Germinal Center/immunology , Humans , Interleukin-2 Receptor beta Subunit/analysis , Mice , NK Cell Lectin-Like Receptor Subfamily A/analysis , T-Lymphocytes, Regulatory/classification , Transforming Growth Factor beta/physiology
14.
Int J Oncol ; 59(5)2021 11.
Article En | MEDLINE | ID: mdl-34533199

Osteosarcoma (OS) is the most common malignant bone tumor and the long­term survival rates remain unsatisfactory. Transforming growth factor­ß (TGF­ß) has been revealed to play a crucial role in OS progression, and RepSox is an effective TGF­ß inhibitor. In the present study, the effect of RepSox on the proliferation of the OS cell lines (HOS and 143B) was detected. The results revealed that RepSox effectively inhibited the proliferation of OS cells by inducing S­phase arrest and apoptosis. Moreover, the inhibitory effect of RepSox on cell migration and invasion was confirmed by wound­healing and Transwell assays. Furthermore, western blotting revealed that the protein levels of molecules associated with the epithelial­mesenchymal transition (EMT) phenotype, including E­cadherin, N­cadherin, Vimentin, matrix metalloproteinase (MMP)­2 and MMP­9, were reduced by RepSox treatment. Concurrently, it was also revealed that the JNK and Smad3 signaling pathway was inhibited. Our in vivo findings using a xenograft model also revealed that RepSox markedly inhibited the growth of tumors. In general, our data demonstrated that RepSox suppressed OS proliferation, EMT and promoted apoptosis by inhibiting the JNK/Smad3 signaling pathway. Thus, RepSox may be a potential anti­OS drug.


Bone Neoplasms/drug therapy , JNK Mitogen-Activated Protein Kinases/physiology , Osteosarcoma/drug therapy , Pyrazoles/pharmacology , Pyridines/pharmacology , Smad3 Protein/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Apoptosis/drug effects , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Osteosarcoma/mortality , Osteosarcoma/pathology , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Signal Transduction/drug effects , Transforming Growth Factor beta/physiology
15.
Int J Oncol ; 59(4)2021 10.
Article En | MEDLINE | ID: mdl-34476496

Epithelial membrane protein 3 (EMP3) is a transmembrane glycoprotein that contains a peripheral myelin protein 22 domain. EMP3 first received attention as a tumor suppressor, but accumulating evidence has since suggested that it may exhibit a tumor­promoting function. Nonetheless, the biological function of EMP3 remains largely unclear with regards to its role in cancer. Herein, it was shown that EMP3 expression is upregulated in non­small cell lung cancer (NSCLC) cells overexpressing aldehyde dehydrogenase 1 (ALDH1). EMP3 was shown to be involved in cell proliferation, the formation of cancer stem cells (CSCs) and in epithelial­mesenchymal transition (EMT). The ability to resist irradiation, one of the characteristics of CSCs, decreased when the EMP3 mRNA expression was knocked down using small interfering RNA. In addition, when EMP3 knockdown reduced the migratory ability of cells, a characteristic of EMT. Additionally, it was shown that the TGF­ß/Smad signaling axis was a target of EMP3. EMP3 was found to interact with TGF­ß receptor type 2 (TGFBR2) upon TGF­ß stimulation in lung CSCs (LCSC). As a result, binding of EMP3­TGFBR2 regulates TGF­ß/Smad signaling activation and consequently affects CSCs and EMT. Kaplan­Meier analysis results confirmed that patients with high expression of EMP3 had poor survival rates. Taken together, these findings showed that EMP3 may be a potential target for management of LCSCs with high expression of ALDH1, and that EMP3 is involved in TGF­ß/Smad signaling activation where it promotes acquisition of cancerous properties in tumors.


Lung Neoplasms/pathology , Membrane Glycoproteins/physiology , Neoplastic Stem Cells/physiology , Transforming Growth Factor beta/physiology , Aldehyde Dehydrogenase 1 Family/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Receptor, Transforming Growth Factor-beta Type II/physiology , Signal Transduction/physiology , Smad Proteins/physiology
16.
Theranostics ; 11(17): 8480-8499, 2021.
Article En | MEDLINE | ID: mdl-34373754

Rationale: Optic neuritis is one of main symptoms in multiple sclerosis (MS) that causes visual disability. Astrocytes are pivotal regulators of neuroinflammation in MS, and astrocytic yes-associated protein (YAP) plays a critical role in neuroinflammation. Meanwhile, YAP signaling is involved in visual impairment, including glaucoma, retinal choroidal atrophy and retinal detachment. However, the roles and underlying mechanisms of astrocytic YAP in neuroinflammation and demyelination of MS-related optic neuritis (MS-ON) remains unclear. Methods: To assess the functions of YAP in MS-ON, experimental autoimmune encephalomyelitis (EAE, a common model of MS) was established, and mice that conditional knockout (CKO) of YAP in astrocytes, YAPGFAP-CKO mice, were successfully generated. Behavior tests, immunostaining, Nissl staining, Hematoxylin-Eosin (HE) staining, TUNEL staining, Luxol Fast Blue (LFB) staining, electron microscopy (EM), quantitative real-time PCR (qPCR), gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) by RNA sequencing were used to examine the function and mechanism of YAP signaling based on these YAPGFAP-CKO mice and EAE model mice. To further explore the potential treatment of YAP signaling in EAE, EAE mice were treated with various drugs, including SRI-011381 that is an agonist of transforming growth factor-ß (TGF-ß) pathway, and XMU-MP-1 which inhibits Hippo kinase MST1/2 to activate YAP. Results: We found that YAP was significantly upregulated and activated in the astrocytes of optic nerve in EAE mice. Conditional knockout of YAP in astrocytes caused more severe inflammatory infiltration and demyelination in optic nerve, and damage of retinal ganglion cells (RGCs) in EAE mice. Moreover, YAP deletion in astrocytes promoted the activation of astrocytes and microglia, but inhibited the proliferation of astrocytes of optic nerve in EAE mice. Mechanically, TGF-ß signaling pathway was significantly down-regulated after YAP deletion in astrocytes. Additionally, both qPCR and immunofluorescence assays confirmed the reduction of TGF-ß signaling pathway in YAPGFAP-CKO EAE mice. Interestingly, SRI-011381 partially rescued the deficits in optic nerve and retina of YAPGFAP-CKO EAE mice. Finally, activation of YAP signaling by XMU-MP-1 relieved the neuroinflammation and demyelination in optic nerve of EAE mice. Conclusions: These results suggest astrocytic YAP may prevent the neuroinflammatory infiltration and demyelination through upregulation of TGF-ß signaling and provide targets for the development of therapeutic strategies tailored for MS-ON.


Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , YAP-Signaling Proteins/metabolism , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Neuroinflammatory Diseases , Optic Nerve/physiology , Optic Neuritis/metabolism , Optic Neuritis/physiopathology , Retina/metabolism , Retina/physiology , Retinal Ganglion Cells/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , YAP-Signaling Proteins/physiology
17.
Cancer Sci ; 112(10): 4198-4207, 2021 Oct.
Article En | MEDLINE | ID: mdl-34375482

Tumor-associated macrophages (TAMs), one of the most common cell components in the tumor microenvironment, have been reported as key contributors to cancer-related inflammation and enhanced metastatic progression of tumors. To explore the underlying mechanism of TAM-induced tumor progression, TAMs were isolated from colorectal cancer patients, and the functional interaction with colorectal cancer cells was analyzed. Our study found that coculture of TAMs contributed to a glycolytic state in colorectal cancer, which promoted the stem-like phenotypes and invasion of tumor cells. TAMs produced the cytokine transforming growth factor-ß to support hypoxia-inducible factor 1α (HIF1α) expression, thereby upregulating Tribbles pseudokinase 3 (TRIB3) in tumor cells. Elevated expression of TRIB3 resulted in activation of the ß-catenin/Wnt signaling pathway, which eventually enhanced the stem-like phenotypes and cell invasion in colorectal cancer. Our findings provided evidence that TAMs promoted colorectal cancer progression in a HIF1α/TRIB3-dependent manner, and blockade of HIF1α signals efficiently improved the outcome of chemotherapy, describing an innovative approach for colorectal cancer treatment.


Cell Cycle Proteins/metabolism , Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/metabolism , Transforming Growth Factor beta/physiology , Tumor-Associated Macrophages/physiology , Animals , Cell Proliferation , Coculture Techniques , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Progression , Female , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplastic Stem Cells , Phenotype , Protein Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism , Up-Regulation , Wnt Signaling Pathway/physiology
18.
PLoS Genet ; 17(7): e1009678, 2021 07.
Article En | MEDLINE | ID: mdl-34260587

Animals can adapt to dynamic environmental conditions by modulating their developmental programs. Understanding the genetic architecture and molecular mechanisms underlying developmental plasticity in response to changing environments is an important and emerging area of research. Here, we show a novel role of cAMP response element binding protein (CREB)-encoding crh-1 gene in developmental polyphenism of C. elegans. Under conditions that promote normal development in wild-type animals, crh-1 mutants inappropriately form transient pre-dauer (L2d) larvae and express the L2d marker gene. L2d formation in crh-1 mutants is specifically induced by the ascaroside pheromone ascr#5 (asc-ωC3; C3), and crh-1 functions autonomously in the ascr#5-sensing ASI neurons to inhibit L2d formation. Moreover, we find that CRH-1 directly binds upstream of the daf-7 TGF-ß locus and promotes its expression in the ASI neurons. Taken together, these results provide new insight into how animals alter their developmental programs in response to environmental changes.


Caenorhabditis elegans Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Adaptation, Physiological/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Cycle , Cell Growth Processes , Cyclic AMP Response Element-Binding Protein/physiology , Gene Expression/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Larva/genetics , Larva/growth & development , Pheromones/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
19.
BMC Nephrol ; 22(1): 203, 2021 05 31.
Article En | MEDLINE | ID: mdl-34059008

INTRODUCTION: The number of patients with end stage kidney disease (ESKD) are increasing world-side. While interstitial fibrosis (IF) is a common step for the progression to ESKD, therapeutic options for IF is still limited in clinical settings. We have reported that bone marrow-derived fibrotic cell, fibrocyte, is involved in the pathogenesis of kidney fibrosis. Also recent studies revealed that erythropoietin has protective effect on kidney diseases. However, it is unknown whether erythropoietin (EPO) inhibits fibrosis in progressive kidney injury. Therefore, we explored the impacts of EPO on kidney fibrosis with focusing on fibrocyte. METHOD: Fibrocyte was differentiated from peripheral mononuclear cells of healthy donor. Fibrocyte was stimulated with transforming growth factor beta (TGF)-ß with/without EPO treatment. Moreover, the therapeutic effect of EPO was evaluated in murine unilateral ureteral obstruction (UUO) model. RESULT: TGF-ß stimulation increased the expression of COL1 mRNA in fibrocyte. EPO signal reduced the expression of COL1 mRNA in dose dependent manner. EPO reduced mitochondrial oxidative stress and ameliorated mitochondrial membrane depolarization induced by TGF-ß stimulation. Moreover, EPO reduced the mRNA expression of mitochondria related molecules, TRAF6, in fibrocyte. In addition, the count of CD45+/αSMA + double-positive fibrocyte was decreased in the EPO-administered UUO kidneys. CONCLUSION: EPO signals function to prevent kidney fibrosis, particularly in fibrocyte. Regulating the renal accumulation of fibrocyte is a part of the anti-fibrotic functions of EPO.


Erythropoietin/physiology , Kidney Diseases/metabolism , Kidney/pathology , Transforming Growth Factor beta/physiology , Animals , Bone Marrow Cells , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Disease Progression , Erythropoietin/therapeutic use , Fibrosis/drug therapy , Fibrosis/metabolism , Humans , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
20.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article En | MEDLINE | ID: mdl-33946990

Ubiquitination and deubiquitination are protein post-translational modification processes that have been recognized as crucial mediators of many complex cellular networks, including maintaining ubiquitin homeostasis, controlling protein stability, and regulating several signaling pathways. Therefore, some of the enzymes involved in ubiquitination and deubiquitination, particularly E3 ligases and deubiquitinases, have attracted attention for drug discovery. Here, we review recent findings on USP15, one of the deubiquitinases, which regulates diverse signaling pathways by deubiquitinating vital target proteins. Even though several basic previous studies have uncovered the versatile roles of USP15 in different signaling networks, those have not yet been systematically and specifically reviewed, which can provide important information about possible disease markers and clinical applications. This review will provide a comprehensive overview of our current understanding of the regulatory mechanisms of USP15 on different signaling pathways for which dynamic reverse ubiquitination is a key regulator.


Protein Processing, Post-Translational/physiology , Signal Transduction/physiology , Ubiquitin-Specific Proteases/physiology , Animals , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/physiology , COP9 Signalosome Complex/physiology , Humans , Immunity, Innate , Male , Mice , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Protein Domains , Protein Isoforms , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Precursors/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Proteases/chemistry , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Wnt Signaling Pathway/physiology , Xenopus Proteins/metabolism
...