RESUMO
Ovarian cancer (OC) is one of the most prevalent and lethal malignancies affecting the female reproductive system, due to its tendency for metastasis and recurrence. This study identified the overexpression of LINC01320 (or long intergenic nonprotein coding RNA 1320) in tissues of ovarian cancer through the analysis of patient samples and online datasets. In vitro and in vivo experiments demonstrate that silencing of LINC01320 expression led to inhibition of proliferation and metastasis of OC cells. RNA pull-down followed by liquid chromatography tandem mass spectrometry (RNA pull-down-LC-MS/MS) revealed that LINC01320 interacted with purine-rich element binding protein B (PURB), a transcriptional repressor. Furthermore, the RNA-seq analysis identified damage-specific DNA binding protein 2 (DDB2) as a major common target of LINC01320 and PURB. Mechanistically, LINC01320 could recruit PURB to the promoter region of DDB2 to repress DDB2 transcription; thus, promoting the expression of NEDD4L and impeding the TGF-ß/SMAD signaling pathway, and ultimately facilitating the progression of OC. Finally, rescue experiments confirmed the involvement of the DDB2/NEDD4L/TGF-ß axis in LINC01320-mediated OC progression. In conclusion, this study unveils for the first time the pivotal function of the LINC01320/PURB/DDB2/NEDD4L/TGF-ß axis and explores its prospective clinical implications in OC.
Assuntos
Movimento Celular , Proliferação de Células , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Ubiquitina-Proteína Ligases Nedd4 , Neoplasias Ovarianas , RNA Longo não Codificante , Fator de Crescimento Transformador beta , Humanos , Feminino , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Movimento Celular/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Linhagem Celular Tumoral , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Ubiquitina-Proteína Ligases Nedd4/genética , Transdução de Sinais , Animais , Camundongos , Camundongos NusRESUMO
Myeloproliferative neoplasms (MPNs) are characterized by an increased production of blood cells due to the acquisition of mutations such as JAK2V617F. TGF-ß, whose secretion is increased in MPN patients, is known to negatively regulate haematopoietic stem cell (HSC) proliferation. Using an isogenic JAK2V617F or JAK2 wild-type UT-7 cell line we observed that JAK2V617F cells resist to TGF-ß antiproliferative activity. Although TGF-ß receptors and SMAD2/3 expressions are similar in both cell types, TGF-ß-induced phosphorylation of SMAD2/3 is reduced in UT-7 JAK2V617F cells compared with JAK2 WT cells. We confirmed that JAK2V617F mutated cells are resistant to the antiproliferative effect of TGF-ß in a competitive assay as we observed a positive selection of JAK2V617F cells when exposed to TGF-ß. Using cell lines, CD34-positive cells from MPN patients and bone marrow cells from JAK2V617F knock-in mice we identified a down regulation of the SHP-1 phosphatase, which is required for the regulation of HSC quiescence by TGF-ß. The transduction of SHP-1 cDNA (but not a phosphatase inactive cDNA) restores the antiproliferative effect of TGF-ß in JAK2V617F mutated cells. Finally, SC-1, a known agonist of SHP-1, antagonized the selection of JAK2V617F mutated cells in the presence of TGF-ß. In conclusion, we show a JAK2-dependent down regulation of SHP-1 in MPN patients' cells which is related to their resistance to the antiproliferative effect of TGF-ß. This may participate in the clonal selection of cancer cells in MPNs.
Assuntos
Proliferação de Células , Janus Quinase 2 , Transtornos Mieloproliferativos , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Fator de Crescimento Transformador beta , Janus Quinase 2/metabolismo , Janus Quinase 2/genética , Humanos , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Camundongos , Linhagem Celular Tumoral , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Regulação para Baixo/genética , Regulação para Baixo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad2/genética , Mutação/genéticaRESUMO
This study aimed to explore the influence and mechanism of low-intensity pulsed ultrasound (LIPUS) combined with Rhodiola bone penetration on the formation of spinal fusion bone. Sixty clean-grade New Zealand white rabbits were selected for randomization and divided into combined group and Rhodiola group, with 30 rabbits in each group to construct a rabbit lumbar intervertebral fusion model, using Rhodiola intervention and Rhodiola combined with LIPUS intervention protocol, respectively. The axial strength, axial stiffness, maximum compressive load, vascular endothelial growth factor (VEGF), cycloxygenase-2 (COX-2), prostaglandin E2 (PGE2) and transforming growth factor-ß (TGF-ß) were compared after HE staining, immunohistochemistry and biomechanical detection. Spine fusion rate was 100.00%; the combined bone graft tissue had implanted bone cell degeneration, cell necrosis and cell hyperplasia, chondrocytes differentiated into trabecular bone and some hematopoietic cells, severe cell necrosis and fiber cell proliferation and late bone formation in the Rhodiola group, VEGF, COX-2, PGE2, TGF-ß, axial strength, axial stiffness, and maximum compression load in the combined group significantly increased (P<0.05). Spinal fusion using LIPUS combined with Rhodiola can enhance biomechanical properties and promote the role of PGE2, COX-2, VEGF, TGF-ß expression and bone formation, and this protocol is worthy of clinical application.
Assuntos
Osteogênese , Rhodiola , Fusão Vertebral , Animais , Coelhos , Rhodiola/química , Osteogênese/efeitos dos fármacos , Fusão Vertebral/métodos , Dinoprostona/metabolismo , Ondas Ultrassônicas , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ciclo-Oxigenase 2/metabolismo , Fenômenos Biomecânicos/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismoRESUMO
Idiopathic pulmonary fibrosis (IPF) is the most common and severe form of pulmonary fibrosis, characterized by scar formation in the lung interstitium. Transforming growth factor beta (TGF-ß) is known as a key mediator in the fibrotic process, acting on fibroblasts and mediating their proliferation and differentiation into myofibroblasts. Although the immune system is not considered responsible for the initiation of IPF, markers of tolerogenic immunity define the pro-fibrotic microenvironment in the lungs. In homeostatic conditions, regulatory T cells (Tregs) constitute the main lymphoid population responsible for maintaining peripheral tolerance. Similar to Tregs, regulatory B cells (Bregs) represent a recently described subset of B lymphocytes with immunosuppressive functions. In the context of IPF, numerous studies have suggested a role for Tregs in enhancing fibrosis, mainly via the secretion of TGF-ß. In humans, most studies show increased percentages of Tregs associated with the severity of IPF, although their exact role remains unclear. In mice, the most commonly used model involves triggering acute lung inflammation with bleomycin, leading to a subsequent fibrotic process. Consequently, data are still conflicting, as Tregs may play a protective role during the inflammatory phase and a deleterious role during the fibrotic phase. Bregs have been less studied in the context of IPF, but their role appears to be protective in experimental models of lung fibrosis. This review presents the latest updates on studies exploring the implication of regulatory lymphoid cells in IPF and compares the different approaches to better understand the origins of conflicting findings.
Assuntos
Linfócitos B Reguladores , Fibrose Pulmonar Idiopática , Linfócitos T Reguladores , Fibrose Pulmonar Idiopática/imunologia , Fibrose Pulmonar Idiopática/patologia , Fibrose Pulmonar Idiopática/metabolismo , Humanos , Linfócitos T Reguladores/imunologia , Animais , Linfócitos B Reguladores/imunologia , Linfócitos B Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Pulmão/imunologia , Pulmão/patologia , Camundongos , Modelos Animais de DoençasRESUMO
Macrophage-myofibroblast transformation (MMT) transforms macrophages into myofibroblasts in a specific inflammation or injury microenvironment. MMT is an essential biological process in fibrosis-related diseases involving the lung, heart, kidney, liver, skeletal muscle, and other organs and tissues. This process consists of interacting with various cells and molecules and activating different signal transduction pathways. This review deeply discussed the molecular mechanism of MMT, clarified crucial signal pathways, multiple cytokines, and growth factors, and formed a complex regulatory network. Significantly, the critical role of transforming growth factor-ß (TGF-ß) and its downstream signaling pathways in this process were clarified. Furthermore, we discussed the significance of MMT in physiological and pathological conditions, such as pulmonary fibrosis and cardiac fibrosis. This review provides a new perspective for understanding the interaction between macrophages and myofibroblasts and new strategies and targets for the prevention and treatment of MMT in fibrotic diseases.
Assuntos
Fibrose , Macrófagos , Miofibroblastos , Transdução de Sinais , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Miofibroblastos/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Fator de Crescimento Transformador beta/metabolismo , Fibrose Pulmonar/patologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/imunologiaRESUMO
This study comprehensively validated the bleomycin (BLEO) induced mouse model of IPF for utility in preclinical drug discovery. To this end, the model was rigorously evaluated for reproducible phenotype and TGFß-directed treatment outcomes. Lung disease was profiled longitudinally in male C57BL6/JRJ mice receiving a single intratracheal instillation of BLEO (n = 10-12 per group). A TGFßR1/ALK5 inhibitor (ALK5i) was profiled in six independent studies in BLEO-IPF mice, randomized/stratified to treatment according to baseline body weight and non-invasive whole-body plethysmography. ALK5i (60 mg/kg/day) or vehicle (n = 10-16 per study) was administered orally for 21 days, starting 7 days after intratracheal BLEO installation. BLEO-IPF mice recapitulated functional, histological and biochemical hallmarks of IPF, including declining expiratory/inspiratory capacity and inflammatory and fibrotic lung injury accompanied by markedly elevated TGFß levels in bronchoalveolar lavage fluid and lung tissue. Pulmonary transcriptome signatures of inflammation and fibrosis in BLEO-IPF mice were comparable to reported data in IPF patients. ALK5i promoted reproducible and robust therapeutic outcomes on lung functional, biochemical and histological endpoints in BLEO-IPF mice. The robust lung fibrotic disease phenotype, along with the consistent and reproducible lung protective effects of ALK5i treatment, makes the spirometry-confirmed BLEO-IPF mouse model highly applicable for profiling novel drug candidates for IPF.
Assuntos
Bleomicina , Modelos Animais de Doenças , Fibrose Pulmonar Idiopática , Camundongos Endogâmicos C57BL , Receptor do Fator de Crescimento Transformador beta Tipo I , Animais , Masculino , Bleomicina/toxicidade , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Camundongos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Espirometria , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/genéticaRESUMO
Ubiquitin C-terminal hydrolase L1 (UCHL1) plays vital roles in cell proliferation, angiogenesis, inflammation and oxidative stress. Nevertheless, it is unclear whether UCHL1 could regulate the biologic behaviour of cells and ultimately influences wound healing. We aim to illustrate the roles and the underlying mechanism of UCHL1 in cutaneous wound healing. Murine full-thickness excisional wound model was utilised to study the effects of UCHL1 on wound healing through topical administration of the UCHL1 inhibitor LDN57444, followed by assessment of wound areas and histological alterations. Subsequently, ethynyldeoxyuridine, scratch and transwell assays were performed to examine fibroblast migration and proliferation. The extracellular matrix (ECM)-related genes expression and transforming growth factor-ß (TGF-ß)/Smad signalling pathways activation were investigated by immuno-fluorescent staining, Western blots and quantitative reverse transcription polymerase chain reaction. We identified elevated UCHL1 expression in non-healing wound tissues. The UCHL1 expression displayed a dynamic change and reached a peak on Day-7 post-wounding during the healing process in mice. Cutaneous administration of LDN57444 promoted wound healing by facilitating collagen deposition, myofibroblast activation and angiogenesis. In vitro experiments demonstrated that UCHL1 concentration dependently inhibited migration, ECM synthesis and activation of human dermal fibroblasts, which was mechanistically related to downregulation of TGF-ß/Smad signalling. Furthermore, these effects could be reversed by TGF-ß inhibitor SB431542. Our findings reveal that UCHL1 is a negative regulator of cutaneous wound healing and considered as a novel prospective therapeutic target for effective wound healing.
Assuntos
Movimento Celular , Fibroblastos , Transdução de Sinais , Proteínas Smad , Fator de Crescimento Transformador beta , Ubiquitina Tiolesterase , Cicatrização , Animais , Humanos , Masculino , Camundongos , Benzamidas/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dioxóis/farmacologia , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/antagonistas & inibidores , Cicatrização/efeitos dos fármacosRESUMO
Metastatic colorectal cancer continues to have a high fatality rate, with approximately only 14% of patients surviving more than 5 years. To improve the survival rate of these patients, the development of new therapeutic drugs is a priority. In this study, we investigated the effects of Oroxylin A on the metastasis of human colorectal cancer cells and its potential molecular mechanism. This study utilised CCK8 assay, transwell assay, flow cytometry, western blot analysis, molecular docking, HE staining, immunofluorescence staining, and xenograft models. The proliferation, migration, and invasion of colon cancer cells were effectively suppressed by Oroxylin A in a dose-dependent manner. Oroxylin A has the potential to inhibit the process of epithelialâmesenchymal transition (EMT) by upregulating the expression of E-cadherin, a marker associated with epithelial cells, while downregulating the levels of N-cadherin, Snail, vimentin, and slug, which are markers associated with mesenchymal cells. In addition, 200 mg/kg of Oroxylin A inhibited the growth of colorectal tumours. Molecular docking technology revealed that Oroxylin A can bind to TGFß and inhibit the activation of the TGFß-smad signalling pathway. The overexpression of TGFß weakened the inhibitory effect of Oroxylin A on the proliferation, migration, and invasion of human colorectal cancer cells, as well as the promoting effect on apoptosis. Oroxylin A inhibited the activation of the TGF-smad signalling pathway and the EMT process, thereby suppressing the migration and invasion of human colorectal cancer cells.
Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Colorretais , Transição Epitelial-Mesenquimal , Flavonoides , Transdução de Sinais , Proteínas Smad , Fator de Crescimento Transformador beta , Humanos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Flavonoides/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Proliferação de Células/efeitos dos fármacos , Camundongos , Movimento Celular/efeitos dos fármacos , Proteínas Smad/metabolismo , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Metástase Neoplásica , Simulação de Acoplamento Molecular , Camundongos NusRESUMO
Increased MMP-9 expression in the tumor microenvironment (TME) plays a crucial role in the extracellular matrix remodeling to facilitate cancer invasion and metastasis. However, the mechanism of MMP-9 upregulation in TME remains elusive. Since TGF-ß and TNF-α levels are elevated in TME, we asked whether these two agents interacted to induce/augment MMP-9 expression. Using a well-established MDA-MB-231 breast cancer model, we found that the synergy between TGF-ß and TNF-α led to MMP-9 upregulation at the transcriptional and translational levels, compared to treatments with each agent alone. Our in vitro findings are corroborated by co-expression of elevated MMP-9 with TGF-ß and TNF-α in human breast cancer tissues. Mechanistically, we found that the MMP-9 upregulation driven by TGF-ß/TNF-α cooperativity was attenuated by selective inhibition of the TGF-ßRI/Smad3 pathway. Comparable outcomes were observed upon inhibition of TGF-ß-induced phosphorylation of Smad2/3 and p38. As expected, the cells defective in Smad2/3 or p38-mediated signaling did not exhibit this synergistic induction of MMP-9. Importantly, the inhibition of histone methylation but not acetylation dampened the synergistic MMP-9 expression. Histone modification profiling further identified the H3K36me2 as an epigenetic regulatory mark of this synergy. Moreover, TGF-ß/TNF-α co-stimulation led to increased levels of the transcriptionally permissive dimethylation mark at H3K36 in the MMP-9 promoter. Comparable outcomes were noted in cells deficient in NSD2 histone methyltransferase. In conclusion, our findings support a cooperativity model in which TGF-ß could amplify the TNF-α-mediated MMP-9 production via chromatin remodeling and facilitate breast cancer invasion and metastasis.
Assuntos
Neoplasias da Mama , Regulação Neoplásica da Expressão Gênica , Metaloproteinase 9 da Matriz , Metástase Neoplásica , Fator de Crescimento Transformador beta , Fator de Necrose Tumoral alfa , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/genética , Fator de Necrose Tumoral alfa/metabolismo , Feminino , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Histonas/metabolismo , Metilação , Transdução de Sinais , Microambiente TumoralAssuntos
Células-Tronco Neoplásicas , Receptor 4 Toll-Like , Fator de Crescimento Transformador beta , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Humanos , Animais , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Camundongos , Fator de Crescimento Transformador beta/metabolismo , Transdução de SinaisRESUMO
The metastasis of hepatocellular carcinoma (HCC) poses a significant threat to the survival of patients. G protein-coupled receptor 56 (GPR56) has garnered extensive attention within malignant tumor research and plays a crucial role in cellular surface signal transmission. Nonetheless, its precise function in HCC remains ambiguous. Our investigation reveals a notable rise in GPR56 expression levels in human HCC cases, with heightened GPR56 levels correlating with unfavorable prognoses. GPR56 regulates TGF-ß pathway by interacting with TGFBR1, thereby promoting HCC metastasis. At the same time, GPR56 is subject to regulation by the canonical cascade of TGF-ß signaling, thereby establishing a positive feedback loop. Furthermore, the combination application of TGFBR1 inhibitor galunisertib (GAL) and GPR56 inhibitor Dihydromunduletone (DHM), significantly inhibits HCC metastasis. Interventions towards this signaling pathway could offer a promising therapeutic approach to effectively impede the metastasis of GPR56-mediated HCC.
Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Metástase Neoplásica , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores Acoplados a Proteínas G , Transdução de Sinais , Fator de Crescimento Transformador beta , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Fator de Crescimento Transformador beta/metabolismo , Animais , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Linhagem Celular Tumoral , Camundongos , Camundongos Nus , Quinolinas/farmacologia , Regulação Neoplásica da Expressão Gênica , Masculino , PirazóisRESUMO
Psychotic and mood disorders are discussed as part of the same continuum. The potential role of immune dysregulation in defining their clinical presentations, however, remains unclear. Differences in TNF-α, IL-6 and TGF-ß levels were investigated in 143 patients with schizophrenia (SCH = 63) and bipolar disorder (BD = 80), in remission. Cytokines were evaluated against the dimensional assessment of psychosis and affective symptoms using the schizo-bipolar scale, together with the severity of the same symptom domains measured by the brief psychiatric rating scale (BPRS). Lower TGF-ß was associated with more lifetime episodes, family risk for psychosis, and more severe mood and psychotic symptoms in all patients. BPRS Affect symptoms domain correlated with lower TGF-ß levels in BD, and higher TGF-ß levels in SCH patients. Using moderated mediation analysis, TGF-ß was a relevant predictor only in the setting of non-categorical symptom distribution, with familial risk for psychosis confirmed as a significant moderator. Severity of BPRS Affect symptoms domain was an independent predictor of inclination towards the psychosis spectrum. The underlying immune dysregulation may be shared by the disorders, rather than a unique characteristic of each, having significant implications for our understanding of the continuum vs. categorical approach to psychosis and mood disorders.
Assuntos
Transtorno Bipolar , Interleucina-6 , Transtornos Psicóticos , Esquizofrenia , Fator de Crescimento Transformador beta , Fator de Necrose Tumoral alfa , Humanos , Feminino , Masculino , Adulto , Fator de Crescimento Transformador beta/sangue , Transtornos Psicóticos/sangue , Interleucina-6/sangue , Fator de Necrose Tumoral alfa/sangue , Esquizofrenia/sangue , Esquizofrenia/imunologia , Transtorno Bipolar/sangue , Transtorno Bipolar/imunologia , Pessoa de Meia-Idade , Afeto , Transtornos do Humor/sangue , Adulto JovemRESUMO
Liver fibrosis is characterized by a wound-healing response and may progress to liver cirrhosis and even hepatocellular carcinoma. Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a tumor suppressor that participates in malignant diseases. However, the role of LHPP in liver fibrosis has not been determined. Herein, the function and regulatory network of LHPP were explored in liver fibrosis. The expression of LHPP in human and murine fibrotic liver tissues was assessed via immunohistochemistry and Western blot analysis. In addition, liver fibrosis was induced in wild-type (WT) and LHPP-/- (KO) mice after carbon tetrachloride (CCl4) or thioacetamide (TAA) treatment. The effect of LHPP was systematically assessed by using specimens acquired from the above murine models. The functional role of LHPP was further explored by detecting the pathway activity of TGF-ß/Smad3 and apoptosis after interfering with LHPP in vitro. To explore whether the function of LHPP depended on the TGF-ß/Smad3 pathway in vivo, an inhibitor of the TGF-ß/Smad3 pathway was used in CCl4-induced WT and KO mice. LHPP expression was downregulated in liver tissue samples from fibrosis patients and fibrotic mice. LHPP deficiency aggravated CCl4- and TAA-induced liver fibrosis. Moreover, through immunoblot analysis, we identified the TGF-ß/Smad3 pathway as a key downstream pathway of LHPP in vivo and in vitro. The effect of LHPP deficiency was reversed by inhibiting the TGF-ß/Smad3 pathway in liver fibrosis. These results revealed that LHPP deficiency exacerbates liver fibrosis through the TGF-ß/Smad3 pathway. LHPP may be a potential therapeutic target in hepatic fibrosis.
Assuntos
Pirofosfatase Inorgânica , Cirrose Hepática , Camundongos Knockout , Transdução de Sinais , Proteína Smad3 , Fator de Crescimento Transformador beta , Animais , Proteína Smad3/metabolismo , Proteína Smad3/genética , Camundongos , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Humanos , Pirofosfatase Inorgânica/metabolismo , Pirofosfatase Inorgânica/genética , Fator de Crescimento Transformador beta/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Apoptose , Tetracloreto de Carbono/toxicidadeRESUMO
We previously demonstrated that the overall number of regulatory T (Treg) cells decrease proportionately with helper CD4+ T cells and their frequencies increase in antiretroviral therapy (ART)-naive human immunodeficiency virus type-1 (HIV-1) infected individuals. The question now is whether the discrepancies in Treg cell numbers and frequencies are synonymous to an impairment of their functions. To address this, we purified Treg cells and assessed their ability to modulate autologous monocytes functions. We observed that Treg cells were able to down modulate autologous monocytes activation as well as interleukin 6 (IL-6) and tumor necrosis factor-alpha (TNF-α) production during stimulation with polyinosinic-polycytidylic acid stabilized with poly-L-lysine and carboxymethylcellulose (poly-ICLC). This activity of Treg cells has been shown to be influenced by immunocompetence including but not limited to helper CD4+ T cell counts, in individuals with HIV-1 infection. Compared to immunosuppressed participants (CD4 < 500 cells/µL), immunocompetent participants (CD4 ≥ 500 cells/µL) showed significantly higher levels of transforming growth factor beta (TGF-ß) and IL-10 (p < 0.001 and p < 0.05, respectively), key cytokines used by Treg cells to exert their immunosuppressive functions. Our findings suggest the contribution of both TGF-ß and IL-10 in the suppressive activity of Treg cells.
Assuntos
Infecções por HIV , HIV-1 , Monócitos , Linfócitos T Reguladores , Humanos , Linfócitos T Reguladores/imunologia , Infecções por HIV/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/fisiologia , Monócitos/imunologia , Masculino , Polilisina/análogos & derivados , Polilisina/farmacologia , Adulto , Poli I-C/imunologia , Poli I-C/farmacologia , Feminino , Pessoa de Meia-Idade , Carboximetilcelulose Sódica/análogos & derivados , Fator de Crescimento Transformador beta/metabolismo , Interleucina-10/metabolismo , Ativação Linfocitária/imunologia , Citocinas/metabolismo , Interleucina-6/metabolismo , Imunocompetência , Fator de Necrose Tumoral alfa/metabolismo , Células CultivadasRESUMO
The synthetic peptide of lumican C-terminal 13 amino acids with the cysteine replaced by an alanine, hereafter referred to as lumikine (LumC13C-A: YEALRVANEVTLN), binds to TGFß type I receptor/activin-like kinase5 (TBR1/ALK5) in the activated TGFß receptor complex to promote corneal epithelial wound healing. The present study aimed to identify the minimum essential amino acid epitope necessary to exert the effects of lumikine via ALK5 and to determine the role of the Y (tyrosine) residue for promoting corneal epithelium wound healing. This study also aimed to determine the signaling pathway(s) triggered by lumican-ALK5 binding. For such, adult Lum knockout (Lum-/-) mice (~8-12 weeks old) were subjected to corneal epithelium debridement using an Agerbrush®. The injured eyes were treated with 10 µL eye drops containing 0.3 µM synthetic peptides designed based on the C-terminal region of lumican for 5-6 h. To unveil the downstream signaling pathways involved, inhibitors of the Alk5 and EGFR signaling pathways were co-administered or not. Corneas isolated from the experimental mice were subjected to whole-mount staining and imaged under a ZEISS Observer to determine the distance of epithelium migration. The expression of EGFR ligands was determined following a scratch assay with HTCE (human telomerase-immortalized cornea epithelial cells) in the presence or not of lumikine. Results indicated that shorter LumC-terminal peptides containing EVTLN and substitution of Y with F in lumikine abolishes its capability to promote epithelium migration indicating that Y and EVTLN are essential but insufficient for Lum activity. Lumikine activity is blocked by inhibitors of Alk5, EGFR, and MAPK signaling pathways, while EGF activity is only suppressed by EGFR and MAPK inhibitors. qRT-PCR of scratched HTCE cells cultures treated with lumikine showed upregulated expression of several EGFR ligands including epiregulin (EREG). Treatment with anti-EREG antibodies abolished the effects of lumikine in corneal epithelium debridement healing. The observations suggest that Lum/lumikine binds Alk5 and promotes the noncanonical Smad-independent TGFß/TBRs signaling pathways during the healing of corneal epithelium debridement.
Assuntos
Epitélio Corneano , Receptores ErbB , Lumicana , Transdução de Sinais , Proteínas Smad , Fator de Crescimento Transformador beta , Cicatrização , Animais , Lumicana/metabolismo , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/metabolismo , Epitélio Corneano/patologia , Cicatrização/efeitos dos fármacos , Receptores ErbB/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Desbridamento , Humanos , Regulação para Cima/efeitos dos fármacos , Ligantes , Camundongos Knockout , Camundongos Endogâmicos C57BLRESUMO
The mechanisms underlying the remarkable capacity of the liver to regenerate are still not completely understood. Particularly, the cross-talk between cytokines and cellular components of the process is of utmost importance because they represent potential avenues for diagnostics and therapeutics. WNT1-inducible-signaling pathway protein 1 (WISP1) is a cytokine member of the CCN family, a family of proteins that play many different roles in liver pathophysiology. WISP1 also belongs to the earliest and strongest upregulated genes in mouse livers after CCl4 intoxication and has recently been shown to be secreted by tumor cells and to bind to type 1 collagen to cause its linearization in vitro and in tumor tissue in vivo. We show that WISP1 expression is strongly induced by TGFß, a critical cytokine in wound healing processes. Additionally, secretion of WISP1 protein by hepatic stellate is increased in cells upon TGFß stimulation (~seven-fold increase). Furthermore, WISP1 facilitates the migration of mouse hepatic stellate cells through collagen in vitro. However, in WISP1 knockout mice, no difference in stellate cell accumulation in damaged liver tissue and no influence on fibrosis was obtained, probably because the knockout of WISP1 was compensated by other factors in vivo.
Assuntos
Proteínas de Sinalização Intercelular CCN , Movimento Celular , Células Estreladas do Fígado , Cirrose Hepática , Camundongos Knockout , Proteínas Proto-Oncogênicas , Fator de Crescimento Transformador beta , Proteínas de Sinalização Intercelular CCN/metabolismo , Proteínas de Sinalização Intercelular CCN/genética , Animais , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Camundongos , Fator de Crescimento Transformador beta/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos Endogâmicos C57BL , MasculinoRESUMO
The peptide hormone kisspeptin attenuates liver steatosis, metabolic dysfunction-associated steatohepatitis (MASH), and fibrosis in mouse models by signaling via the kisspeptin 1 receptor (KISS1R). However, whether kisspeptin impacts fibrogenesis in the human liver is not known. We investigated the impact of a potent kisspeptin analog (KPA) on fibrogenesis using human precision-cut liver slices (hPCLS) from fibrotic livers from male patients, in human hepatic stellate cells (HSCs), LX-2, and in primary mouse HSCs. In hPCLS, 48 h and 72 h of KPA (3 nM, 100 nM) treatment decreased collagen secretion and lowered the expression of fibrogenic and inflammatory markers. Immunohistochemical studies revealed that KISS1R is expressed and localized to HSCs in MASH/fibrotic livers. In HSCs, KPA treatment reduced transforming growth factor b (TGFß)-the induced expression of fibrogenic and inflammatory markers, in addition to decreasing TGFß-induced collagen secretion, cell migration, proliferation, and colony formation. Mechanistically, KISS1R signaling downregulated TGFß signaling by decreasing SMAD2/3 phosphorylation via the activation of protein phosphatases, PP2A, which dephosphorylates SMAD 2/3. This study revealed for the first time that kisspeptin reverses human hepatic fibrogenesis, thus identifying it as a new therapeutic target to treat hepatic fibrosis.
Assuntos
Células Estreladas do Fígado , Kisspeptinas , Cirrose Hepática , Transdução de Sinais , Fator de Crescimento Transformador beta , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , Kisspeptinas/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Animais , Masculino , Camundongos , Receptores de Kisspeptina-1/metabolismo , Receptores de Kisspeptina-1/genética , Proliferação de Células/efeitos dos fármacos , Proteína Smad2/metabolismo , Movimento Celular/efeitos dos fármacosRESUMO
In idiopathic pulmonary fibrosis (IPF), epithelial abnormalities are present including bronchiolization and alveolar cell dysfunction. We hypothesized that the IPF microenvironment disrupts normal epithelial growth and differentiation. We mimicked the soluble factors within an IPF microenvironment using an IPF cocktail (IPFc), composed of nine factors which are increased in IPF lungs (CCL2, IL-1ß, IL-4, IL-8, IL-13, IL-33, TGF-ß, TNFα, and TSLP). Using IPFc, we asked whether the soluble factor milieu in IPF affects epithelial growth and differentiation and how IPFc compares to TGF-ß alone. Epithelial growth and differentiation were studied using mouse lung organoids (primary Epcam+ epithelial cells co-cultured with CCL206 fibroblasts). Organoids exposed to IPFc and TGF-ß were re-sorted into epithelial and fibroblast fractions and subjected to RNA sequencing. IPFc did not affect the number of organoids formed. However, pro-surfactant protein C expression was decreased. On these parameters, TGF-ß alone had similar effects. However, RNA sequencing of re-sorted organoids revealed that IPFc and TGF-ß had distinct effects on both epithelial cells and fibroblasts. IPFc upregulated goblet cell markers, whereas these were inhibited by TGF-ß. Although both IPFc and TGF-ß increased extracellular matrix gene expression, only TGF-ß increased myofibroblast markers. VEGF-C and Wnt signaling were among the most differentially regulated signaling pathways by IPFc versus TGF-ß. Interestingly, Wnt pathway activation rescued Sftpc downregulation induced by IPFc. In conclusion, IPFc alters epithelial differentiation in a way that is distinct from TGF-ß. Alterations in Wnt signaling contribute to these effects. IPFc may be a more comprehensive representation of the soluble factor microenvironment in IPF.
Assuntos
Diferenciação Celular , Células Epiteliais , Fibrose Pulmonar Idiopática , Fator de Crescimento Transformador beta , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Animais , Camundongos , Células Epiteliais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Humanos , Organoides/metabolismo , Organoides/patologia , Pulmão/metabolismo , Pulmão/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Via de Sinalização Wnt , Camundongos Endogâmicos C57BL , Células CultivadasRESUMO
At the ASCO 2024 meeting, Anthony P Conley, coauthor on this editorial, presented promising data from the phase 1/2 clinical trial called BETA PRIME (ClinicalTrials.gov NCT04673942) with AdAPT-001 plus a checkpoint inhibitor (CI). All participants gave informed consent to participate in BETA PRIME before taking part. AdAPT-001 is an oncolytic adenovirus that expresses a transforming growth factor beta (TGF-ß) trap to neutralize active TGF-ß. This editorial proposes that the TGF-ß trap of AdAPT-001 reverses the immunosuppressive environment of tumor cells, and thus makes these tumors susceptible to CIs like the anti-PD-1 agent, nivolumab, and potentially other therapies as well.
Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Fator de Crescimento Transformador beta , Humanos , Resistencia a Medicamentos Antineoplásicos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Fator de Crescimento Transformador beta/metabolismo , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como AssuntoRESUMO
Abnormal autophagy and the transforming growth factor-ß (TGFß)-SMAD3/7 signaling pathway play an important role in the development of intrauterine adhesions (IUAs); however, the exact underlying mechanisms remain unclear. In this study, we used IUA patient tissue and SMAD7 conditional knockout mice to detect whether SMAD7 effected IUA via regulation of autophagy and the TGFß-SMAD3 signaling pathway. We applied a combination of techniques for the detection of p-SMAD3, SMAD7, autophagy and fibrosis-related proteins, autophagic flux, and analysis of the SMAD3 binding site. Endometrial tissue of patients with IUA exhibited lower expression levels of SMAD7. In endometrial stromal cells, silencing of SMAD7 inhibited autophagic flux, whereas overexpressed SMAD7 promoted autophagic flux. This SMAD7-mediated autophagic flux regulates the stromal-myofibroblast transition, and these phenotypes were regulated by the TGFß-SMAD3 signaling pathway. SMAD3 directly binds to the 3'-untranslated region of transcription factor EB (TFEB) and inhibits its transcription. SMAD7 promoted autophagic flux by inhibiting SMAD3, thereby promoting the expression of TFEB. In SMAD7 conditional knockout mice, the endometria showed a fibrotic phenotype. Simultaneously, autophagic flux was inhibited. On administering the autophagy activator rapamycin, this endometrial fibrosis phenotype was partially reversed. The loss of SMAD7 promotes endometrial fibrosis by inhibiting autophagic flux via the TGFß-SMAD3 pathway. Therefore, this study reveals a potential therapeutic target for IUA.