Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.249
Filtrar
1.
Iran J Immunol ; 19(2): 184-192, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35767891

RESUMO

BACKGROUND: Concomitant signals from IL-6 and TGF-ß have a central role in the Th17 cells development and differentiation, and these cells are the main promoters of demyelinating inflammation in the central nervous system (CNS) resulting in multiple sclerosis (MS). OBJECTIVES: To evaluate the simultaneous IL-6 and TGF-ß gene and their receptor protein expression in patients with Relapsing-Remitting (RR)-MS. MATERIALS AND METHODS: IL-6 and TGF-ß mRNA and their receptor expression on the surface of CD4+T cells were evaluated using real-time PCR (RT-PCR) and flow cytometry, respectively. RESULTS: The IL-6 mRNA expression in patients with RRMS was significantly higher than in the controls (p= 0.019). When patients who did not receive any other treatment were compared with the controls, the significant difference was substantial (p=0.006). The TGF-ß mRNA expression in patients was lower than in the controls (p = 0.03). However, in patients receiving IFNß, it increased compared with the other patients (p= 0.036). There was no difference in cytokine receptor expression between patients and the control group. CONCLUSION: Our data conclude an increase and decrease in mRNA expression levels of IL-6 and TGF-ß in patients with RRMS, respectively. Moreover, there were no significant differences in receptor expression of either cytokines. Based on our data the balance of TGF and IL-6 appears to have a positive impact on the disease control.


Assuntos
Interferon beta , Interleucina-6 , Esclerose Múltipla Recidivante-Remitente , Fator de Crescimento Transformador beta , Citocinas/biossíntese , Citocinas/sangue , Citocinas/genética , Humanos , Interferon beta/genética , Interferon beta/farmacologia , Interleucina-6/análogos & derivados , Interleucina-6/biossíntese , Interleucina-6/sangue , Interleucina-6/genética , Esclerose Múltipla Recidivante-Remitente/sangue , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/sangue , Fator de Crescimento Transformador beta/genética
2.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35105806

RESUMO

The protumor roles of alternatively activated (M2) tumor-associated macrophages (TAMs) have been well established, and macrophage reprogramming is an important therapeutic goal. However, the mechanisms of TAM polarization remain incompletely understood, and effective strategies for macrophage targeting are lacking. Here, we show that miR-182 in macrophages mediates tumor-induced M2 polarization and can be targeted for therapeutic macrophage reprogramming. Constitutive miR-182 knockout in host mice and conditional knockout in macrophages impair M2-like TAMs and breast tumor development. Targeted depletion of macrophages in mice blocks the effect of miR-182 deficiency in tumor progression while reconstitution of miR-182-expressing macrophages promotes tumor growth. Mechanistically, cancer cells induce miR-182 expression in macrophages by TGFß signaling, and miR-182 directly suppresses TLR4, leading to NFκb inactivation and M2 polarization of TAMs. Importantly, therapeutic delivery of antagomiR-182 with cationized mannan-modified extracellular vesicles effectively targets macrophages, leading to miR-182 inhibition, macrophage reprogramming, and tumor suppression in multiple breast cancer models of mice. Overall, our findings reveal a crucial TGFß/miR-182/TLR4 axis for TAM polarization and provide rationale for RNA-based therapeutics of TAM targeting in cancer.


Assuntos
Reprogramação Celular , Neoplasias Mamárias Animais/metabolismo , MicroRNAs/metabolismo , RNA Neoplásico/metabolismo , Transdução de Sinais , Macrófagos Associados a Tumor/metabolismo , Animais , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , RNA Neoplásico/genética , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/genética , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética
3.
Mol Cell Biochem ; 477(1): 53-65, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34533647

RESUMO

The era of induced pluripotent stem cells (iPSCs) was used as novel biotechnology to replace embryonic stem cells bypassing the ethical concerns and problems of stem cell transplant rejection. The anti-tumour potential of iPSCs against many tumours including salivary cancer was proven in previous studies. The current study aimed to investigate the contribution of the Bax, Sirt-1, TGF-ß, and MALAT genes and/or their protein expression to the pathogenesis of submandibular carcinogenesis before and after iPSCs treatment. Thirty Wistar albino rats were equally assigned into three groups: group I (control), group II (Squamous cell carcinoma (SCC)): submandibular glands were injected SCC cells, and group III (SCC/iPSCs): SCC rats were treated by 5 × 106 iPSCs. Submandibular gland sections were subjected to histological and immunohistochemical analyses to detect mucopolysaccharides, Bax, and TGF-ß expression as well as PCR quantification for TGF-ß, Sirt-1, and lncRNA MALAT-1 gene expressions. Western blotting was also used to detect Sirt-1 and TGF-ß protein expressions. SCC group revealed infiltration by sheets of malignant squamous cells with or without keratin pearls and inflammatory cells, in addition to upregulation of TGF-ß, Sirt-1, MALAT-1, and Bax, whereas SCC/iPSCs group showed an improved submandibular histoarchitecture with the maintenance of the secretory function. Bax and TGF-ß immunoexpression were significantly reduced. The upregulated TGF-ß, Sirt-1, and MALAT-1 genes were significantly decreased. iPSCs protected against the experimentally induced submandibular gland carcinoma that might be achieved via their regenerative potential and their regulatory modulation of Sirt-1, TGF-ß, and MALAT-1 gene/protein expressions and of the apoptotic response in cancer cells.


Assuntos
Apoptose , Carcinoma de Células Escamosas , Células-Tronco Pluripotentes Induzidas , RNA Longo não Codificante/biossíntese , RNA Neoplásico/biossíntese , Neoplasias das Glândulas Salivares , Sirtuína 1/biossíntese , Glândula Submandibular/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/terapia , Linhagem Celular Tumoral , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Masculino , Ratos , Ratos Wistar , Neoplasias das Glândulas Salivares/metabolismo , Neoplasias das Glândulas Salivares/terapia , Proteína X Associada a bcl-2/biossíntese
4.
Respir Res ; 22(1): 315, 2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930252

RESUMO

Repetitive aeroallergen exposure is linked to sensitization and airway remodeling through incompletely understood mechanisms. In this study, we examine the dynamic mucosal response to cat dander extract (CDE), a ubiquitous aero-allergen linked to remodeling, sensitization and asthma. We find that daily exposure of CDE in naïve C57BL/6 mice activates innate neutrophilic inflammation followed by transition to a lymphocytic response associated with waves of mucosal transforming growth factor (TGF) isoform expression. In parallel, enhanced bronchiolar Smad3 expression and accumulation of phospho-SMAD3 was observed, indicating paracrine activation of canonical TGFßR signaling. CDE exposure similarly triggered epithelial cell plasticity, associated with expression of mesenchymal regulatory factors (Snai1 and Zeb1), reduction of epithelial markers (Cdh1) and activation of the NFκB/RelA transcriptional activator. To determine whether NFκB functionally mediates CDE-induced growth factor response, mice were stimulated with CDE in the absence or presence of a selective IKK inhibitor. IKK inhibition substantially reduced the level of CDE-induced TGFß1 expression, pSMAD3 accumulation, Snai1 and Zeb1 expression. Activation of epithelial plasticity was demonstrated by flow cytometry in whole lung homogenates, where CDE induces accumulation of SMA+Epcam+ population. Club cells are important sources of cytokine and growth factor production. To determine whether Club cell innate signaling through NFκB/RelA mediated CDE induced TGFß signaling, we depleted RelA in Secretoglobin (Scgb1a1)-expressing bronchiolar cells. Immunofluorescence-optical clearing light sheet microscopy showed a punctate distribution of Scgb1a1 progenitors throughout the small airway. We found that RelA depletion in Secretoglobin+ cells results in inhibition of the mucosal TGFß response, blockade of EMT and reduced subepithelial myofibroblast expansion. We conclude that the Secretoglobin-derived bronchiolar cell is central to coordinating the innate response required for mucosal TGFß1 response, EMT and myofibroblast expansion. These data have important mechanistic implications for how aero-allergens trigger mucosal injury response and remodeling in the small airway.


Assuntos
Remodelação das Vias Aéreas , Asma/genética , Regulação da Expressão Gênica , Miofibroblastos/metabolismo , NF-kappa B/genética , Secretoglobinas/metabolismo , Fator de Crescimento Transformador beta/genética , Alérgenos/efeitos adversos , Animais , Asma/metabolismo , Asma/patologia , Bronquíolos/metabolismo , Bronquíolos/patologia , Gatos , Transdiferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/patologia , NF-kappa B/biossíntese , Transdução de Sinais , Fator de Crescimento Transformador beta/biossíntese
5.
Front Immunol ; 12: 746187, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34616408

RESUMO

Regulatory B cells (Bregs) have an anti-inflammatory role and can suppress autoimmunity, by employing both cytokine secretion and cell-contact mediated mechanisms. Numerous Breg subsets have been described and have overlapping phenotypes in terms of their immune expression markers or cytokine production. A hallmark feature of Bregs is the secretion of IL-10, although IL-35 and TGFß-producing B cells have also been identified. To date, few reports have identified an impaired frequency or function of Bregs in individuals with type 1 diabetes; thus our understanding of the role played by these Breg subsets in the pathogenesis of this condition is limited. In this review we will focus on how regulatory B cells are altered in the development of type 1 diabetes, highlighting both frequency and function and discuss both human and animal studies.


Assuntos
Linfócitos B Reguladores/imunologia , Diabetes Mellitus Tipo 1/imunologia , Adolescente , Adulto , Animais , Autoimunidade , Feminino , Humanos , Imunofenotipagem , Interleucina-10/biossíntese , Interleucinas/biossíntese , Ilhotas Pancreáticas/imunologia , Masculino , Camundongos , Fator de Crescimento Transformador beta/biossíntese , Adulto Jovem
6.
Front Immunol ; 12: 717370, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489969

RESUMO

An appropriate development of the placenta consisting of trophoblast cell migration, invasion, proliferation, and apoptosis, is essential to establishing and maintaining a successful pregnancy. Ubiquitin-specific protease 2a (USP2a) regulates the processes of metastasis in multiple tumor cells. Yet, no known research has focused on exploring the effect of USP2a on trophoblasts and its possible mechanism in the pathogenies of recurrent miscarriage (RM). In this study, we first detected the decreased mRNA levels and the protein levels of USP2a in placental villous tissue samples from the RM patients. In vitro assays verified that overexpression of USP2a promoted human trophoblast proliferation, migration, invasion, whereas knockdown of USP2a inhibited these processes. Mechanistically, USP2a activated PI3K/Akt/GSK3ß signaling pathway to promote nuclear translocation of ß-catenin and further activated epithelial-mesenchymal transition (EMT) in the trophoblasts. Moreover, transforming growth factor-beta (TGF-ß) up-regulated USP2a expression in trophoblasts. Interestingly, M2 macrophage secreted TGF-ß induced trophoblast migration and invasion, and an anti-TGF-ß antibody alleviated this effect. Collectively, this study indicated that USP2a regulated trophoblast invasion and that abnormal USP2a expression might lead to aberrant trophoblast invasion, thus contributing to RM.


Assuntos
Aborto Habitual/etiologia , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Trofoblastos/metabolismo , Ubiquitina Tiolesterase/genética , Aborto Habitual/metabolismo , Biomarcadores , Movimento Celular , Proliferação de Células , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Macrófagos/imunologia , Macrófagos/metabolismo , Placenta/imunologia , Placenta/metabolismo , Gravidez , Transporte Proteico , Fator de Crescimento Transformador beta/biossíntese , Ubiquitina Tiolesterase/metabolismo
7.
Front Immunol ; 12: 716939, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34434198

RESUMO

Conjunctival epithelium forms a barrier between the ocular surface microbial flora and the ocular mucosa. In addition to secreting gel-forming mucins, goblet cells, located in the conjunctival epithelium, help maintain local immune homeostasis by secreting active TGFß2 and promoting tolerogenic phenotype of dendritic cells in the vicinity. Although dendritic cell subsets, characteristic of mucosal tissues, are found in the conjunctiva, previous studies provided limited information about their location within the tissue. In this study, we examine immunostained conjunctiva explants to determine the location of CD11c-positive dendritic cells in the context of MUC5AC-positive goblet cells. Considering that conjunctival goblet cells are responsive to signaling induced by pathogen recognition receptors, we also assess if their responses to microbial product, flagellin, can contribute to the disruption of ocular mucosal homeostasis that promotes activation of dendritic cells and results in chronic ocular surface inflammation. We find that dendritic cells in the conjunctiva with an increased microbial colonization are located adjacent to goblet cells. While their cell bodies in the stromal layer are immediately below the epithelial layer, several extensions of dendritic cells are projected across the epithelium towards the ocular surface. Such trans-epithelial dendrites are not detectable in healthy ocular mucosa. In response to topically applied flagellin, increased proportion of CD11c-positive cells in the conjunctiva strongly express MHC class II relative to the untreated conjunctiva. This change is accompanied by reduced immunoreactivity to TGFß-activating Thrombospondin-1 in the conjunctival epithelium. These findings are supported by in vitro observations in primary cultures of goblet cells that respond to the TLR5 stimulation with an increased expression of IL-6 and reduced level of active TGFß. The observed changes in the conjunctiva after flagellin application correspond with the development of clinical signs of chronic ocular mucosal inflammation including corneal epitheliopathy. Collectively, these findings demonstrate the ability of ocular mucosal dendritic cells to extend trans-epithelial dendrites in response to increased microbial colonization at the ocular surface. Moreover, this study provides key insight into how goblet cell responses to microbial stimuli may contribute to the disruption of ocular mucosal homeostasis and chronic ocular mucosal inflammation.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Túnica Conjuntiva/imunologia , Túnica Conjuntiva/metabolismo , Células Caliciformes/metabolismo , Receptor 5 Toll-Like/metabolismo , Animais , Apresentação de Antígeno , Biomarcadores , Comunicação Celular/imunologia , Células Cultivadas , Túnica Conjuntiva/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Imunofluorescência , Expressão Gênica , Células Caliciformes/imunologia , Homeostase , Imunomodulação , Camundongos , Camundongos Knockout , Mucosa/citologia , Mucosa/imunologia , Mucosa/metabolismo , Fator de Crescimento Transformador beta/biossíntese
8.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34265069

RESUMO

Cancer-induced skeletal muscle defects show sex-specific differences in severity with men performing poorly compared to women. Hormones and sex chromosomal differences are suggested to mediate these differences, but the functional skeletal muscle markers to document these differences are unknown. We show that the myogenic microRNA miR-486 is a marker of sex-specific differences in cancer-induced skeletal muscle defects. Cancer-induced loss of circulating miR-486 was more severe in men with bladder, lung, and pancreatic cancers compared to women with the same cancer types. In a syngeneic model of pancreatic cancer, circulating and skeletal muscle loss of miR-486 was more severe in male mice compared to female mice. Estradiol (E2) and the clinically used selective estrogen receptor modulator toremifene increased miR-486 in undifferentiated and differentiated myoblast cell line C2C12 and E2-inducible expression correlated with direct binding of estrogen receptor alpha (ERα) to the regulatory region of the miR-486 gene. E2 and toremifene reduced the actions of cytokines such as myostatin, transforming growth factor ß, and tumor necrosis factor α, which mediate cancer-induced skeletal muscle wasting. E2- and toremifene-treated C2C12 myoblast/myotube cells contained elevated levels of active protein kinase B (AKT) with a corresponding decrease in the levels of its negative regulator PTEN, which is a target of miR-486. We propose an ERα:E2-miR-486-AKT signaling axis, which reduces the deleterious effects of cancer-induced cytokines/chemokines on skeletal muscle mass and/or function.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/biossíntese , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Neoplasias/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Estradiol/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Doenças Musculares/complicações , Miostatina/biossíntese , Neoplasias/complicações , Fatores Sexuais , Transdução de Sinais , Toremifeno/farmacologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Necrose Tumoral alfa/biossíntese
9.
J Am Heart Assoc ; 10(11): e019276, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34041919

RESUMO

Background Aortic dissection (AD) is one of the most life-threatening cardiovascular diseases that exhibit high genetic heterogeneity. However, it is unclear whether variants within the COL5A1 gene can cause AD. Therefore, we intend to determine whether COL5A1 is a causative gene of AD. Methods and Results We performed targeted sequencing in 702 patients with unrelated sporadic AD and 163 matched healthy controls using a predesigned panel with 152 vessel matrix-related genes. As a result, we identified that 11 variants in COL5A1 caused AD in 11 out of the 702 patients with AD. Furthermore, Col5a1 knockout (Col5a1+/-) rats were generated through the CRISPR/Cas9 system. Although there was no spontaneous AD, electron microscopy revealed a fracture of elastic fibers and disarray of collagenous fibers in 6-week-old Col5a1+/- rats, but not in WT rats (93.3% versus 0.0%, P<0.001). Three-week-old rats were used to induce the AD phenotype with ß-aminopropionitrile monofumarate for 4 weeks followed by angiotensin II for 72 hours. The ß-aminopropionitrile monofumarate and angiotensin II-treated rat model confirmed that Col5a1+/- rats had considerably higher AD incidence than WT rats. Subsequent mechanism analyses demonstrated that the transforming growth factor-ß-signaling pathway was significantly activated in Col5a1+/- rats. Conclusions Our findings, for the first time, revealed a relationship between variants in COL5A1 and AD via targeted sequencing in 1.57% patients with sporadic aortic dissection. The Col5a1 knockout rats exhibited AD after an intervention, indicating that COL5A1 is a causative gene of AD. Activation of the transforming growth factor-ß-signaling pathway may be implicated in the pathogenesis of this kind of AD.


Assuntos
Aneurisma da Aorta Torácica/genética , Dissecção Aórtica/genética , Colágeno Tipo V/genética , Fator de Crescimento Transformador beta/genética , Dissecção Aórtica/diagnóstico , Dissecção Aórtica/metabolismo , Animais , Aorta Torácica/ultraestrutura , Aneurisma da Aorta Torácica/diagnóstico , Aneurisma da Aorta Torácica/metabolismo , Western Blotting , Colágeno Tipo V/biossíntese , DNA/genética , Modelos Animais de Doenças , Feminino , Seguimentos , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Microscopia Eletrônica , Pessoa de Meia-Idade , Fenótipo , Ratos , Ratos Transgênicos , Estudos Retrospectivos , Transdução de Sinais , Tomografia Computadorizada por Raios X , Fator de Crescimento Transformador beta/biossíntese
10.
Exp Eye Res ; 209: 108629, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34029595

RESUMO

The shortage of human donor corneas has raised important concerns about engineering of corneal endothelial cells (CECs) for clinical use. However, due to the limited proliferative capacity of human CECs, driving them into proliferation and regeneration may be difficult. Unlike human CECs, rabbit CECs have a marked proliferative capacity. To clarify the potential reason for this difference, we analysed the proteomes of four human corneal endothelium samples and four rabbit corneal endothelium samples with quantitative label-free proteomics and downstream analysis. We discovered that vitamin and selenocompound metabolism and some signaling pathways such as NF-kappa B signaling pathway differed between the samples. Moreover, TGFß, PITX2 and keratocan were distinctively expressed in rabbit samples, which might be associated with active proliferation in rabbit CECs. This study illustrates the proteomic differences between human and rabbit CECs and might promote CEC engineering strategies.


Assuntos
Doenças da Córnea/metabolismo , Transplante de Córnea/métodos , Endotélio Corneano/metabolismo , Proteínas de Homeodomínio/biossíntese , Proteoma/metabolismo , Proteômica/métodos , Fatores de Transcrição/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Idoso , Animais , Diferenciação Celular , Células Cultivadas , Doenças da Córnea/patologia , Doenças da Córnea/cirurgia , Modelos Animais de Doenças , Endotélio Corneano/citologia , Endotélio Corneano/transplante , Humanos , Masculino , Pessoa de Meia-Idade , Coelhos , Preservação de Tecido/métodos
11.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805788

RESUMO

Persistent chronic liver diseases increase the scar formation and extracellular matrix accumulation that further progress to liver fibrosis and cirrhosis. Nevertheless, there is no antifibrotic therapy to date. The ketogenic diet is composed of high fat, moderate to low-protein, and very low carbohydrate content. It is mainly used in epilepsy and Alzheimer's disease. However, the effects of the ketogenic diet on liver fibrosis remains unknown. Through ketogenic diet consumption, ß-hydroxybutyrate (bHB) and acetoacetate (AcAc) are two ketone bodies that are mainly produced in the liver. It is reported that bHB and AcAc treatment decreases cancer cell proliferation and promotes apoptosis. However, the influence of bHB and AcAc in hepatic stellate cell (HSC) activation and liver fibrosis are still unclear. Therefore, this study aimed to investigate the effect of the ketogenic diet and ketone bodies in affecting liver fibrosis progression. Our study revealed that feeding a high-fat ketogenic diet increased cholesterol accumulation in the liver, which further enhanced the carbon tetrachloride (CCl4)- and thioacetamide (TAA)-induced liver fibrosis. In addition, more severe liver inflammation and the loss of hepatic antioxidant and detoxification ability were also found in ketogenic diet-fed fibrotic mouse groups. However, the treatment with ketone bodies (bHB and AcAc) did not suppress transforming growth factor-ß (TGF-ß)-induced HSC activation, platelet-derived growth factor (PDGF)-BB-triggered proliferation, and the severity of CCl4-induced liver fibrosis in mice. In conclusion, our study demonstrated that feeding a high-fat ketogenic diet may trigger severe steatohepatitis and thereby promote liver fibrosis progression. Since a different ketogenic diet composition may exert different metabolic effects, more evidence is necessary to clarify the effects of a ketogenic diet on disease treatment.


Assuntos
Ácido 3-Hidroxibutírico/farmacologia , Acetoacetatos/farmacologia , Colesterol/biossíntese , Dieta Cetogênica/efeitos adversos , Cirrose Hepática/metabolismo , Fígado/efeitos dos fármacos , Ácido 3-Hidroxibutírico/biossíntese , Acetoacetatos/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Becaplermina/farmacologia , Tetracloreto de Carbono/administração & dosagem , Catalase/genética , Catalase/metabolismo , Proliferação de Células/efeitos dos fármacos , Colesterol/sangue , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Desmina/genética , Desmina/metabolismo , Progressão da Doença , Regulação da Expressão Gênica/efeitos dos fármacos , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Índice de Gravidade de Doença , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Tioacetamida/administração & dosagem , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/farmacologia
12.
Neurotherapeutics ; 18(3): 1963-1979, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33860461

RESUMO

Adult neurogenesis is a target for brain rejuvenation as well as regeneration in aging and disease. Numerous approaches showed efficacy to elevate neurogenesis in rodents, yet translation into therapies has not been achieved. Here, we introduce a novel human TGFß-RII (Transforming Growth Factor-Receptor Type II) specific LNA-antisense oligonucleotide ("locked nucleotide acid"-"NVP-13"), which reduces TGFß-RII expression and downstream receptor signaling in human neuronal precursor cells (ReNcell CX® cells) in vitro. After we injected cynomolgus non-human primates repeatedly i.th. with NVP-13 in a preclinical regulatory 13-week GLP-toxicity program, we could specifically downregulate TGFß-RII mRNA and protein in vivo. Subsequently, we observed a dose-dependent upregulation of the neurogenic niche activity within the hippocampus and subventricular zone: human neural progenitor cells showed significantly (up to threefold over control) enhanced differentiation and cell numbers. NVP-13 treatment modulated canonical and non-canonical TGFß pathways, such as MAPK and PI3K, as well as key transcription factors and epigenetic factors involved in stem cell maintenance, such as MEF2A and pFoxO3. The latter are also dysregulated in clinical neurodegeneration, such as amyotrophic lateral sclerosis. Here, we provide for the first time in vitro and in vivo evidence for a novel translatable approach to treat neurodegenerative disorders by modulating neurogenesis.


Assuntos
Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Esclerose Amiotrófica Lateral/metabolismo , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Macaca fascicularis , Masculino , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Primatas , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/biossíntese
13.
Sci Rep ; 11(1): 6054, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33723310

RESUMO

Resveratrol is a phytochemical with anti-angiogenic, anti-inflammatory, and antioxidant properties. The present study has evaluated the effect of resveratrol on the expression of vascular endothelial growth factor (VEGF), transforming growth factor-ß (TGF-ß) and matrix metalloproteinase-9 (MMP-9) as factors related to endometriosis progression. Thirteen eutopic (EuESCs) and 8 ectopic (EESCs) endometrial stromal cells from women with endometriosis and 11 control endometrial stromal cells (CESCs) were treated with resveratrol (100 µM) for 6, 24 and 48 h. The gene and protein expression levels of VEGF, TGF-ß, and MMP-9 were measured using real-time PCR and ELISA methods, respectively. Results showed that the basal gene and protein expression of VEGF and MMP-9 were higher in EESCs compared to EuESCs and CESCs (P < 0.01 to < 0.001 and P < 0.05 to < 0.01 respectively). Also, resveratrol treatment decreased the gene and protein expression of VEGF and MMP-9 in EuESCs, EESCs and CESCs (P < 0.05 to < 0.01 and P < 0.05 to < 0.01 respectively) and gene and protein expression of TGF-ß in EESCs and EuESCs (P < 0.05 to < 0.01). The effect of resveratrol in reduction of VEGF gene expression was statistically more noticeable in EESCs compared to EuESCs and CESCs (P < 0.05). According to the findings, resveratrol may ameliorate endometriosis progression through reducing the expression of VEGF, TGF-ß, and MMP-9 in endometrial stromal cells (ESCs).


Assuntos
Endometriose/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Metaloproteinase 9 da Matriz/biossíntese , Resveratrol/farmacologia , Fator de Crescimento Transformador beta/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adulto , Células Cultivadas , Relação Dose-Resposta a Droga , Endometriose/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Células Estromais/metabolismo , Células Estromais/patologia
14.
Biochimie ; 185: 117-127, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33771655

RESUMO

Fibrosis is a pathological process in diabetic nephropathy that causes renal failure and dysfunction. Given the known anti-diabetic effects of trans-Anethole (TA), we aimed to investigate its renoprotective and anti-fibrotic effect alone and in combination with losartan in diabetic nephropathy. Male Wistar rats received a single intraperitoneal injection of 65 mg/kg streptozotocin (STZ) for diabetes induction. Diabetic rats were treated orally with saline, TA (80 mg/kg), losartan (Los; 10 mg/kg), or the combination of TA and losartan (TA-Los) daily for five weeks. Renal function was monitored during the study, and renal fibrosis, oxidative stress markers, apoptotic cells, and the expression and localization of AT1R, TGF-ß1, and Col-IV were detected in the kidney. Results showed that TA alone and in combination with losartan was able to decrease blood glucose, urea, and creatinine levels and improve kidney function parameters. TA, Los, and TA-Los significantly reduced tubule vascular degeneration, glomerular and tubulointerstitial sclerosis, oxidative stress, and apoptotic cells. Immunohistochemistry analyses showed that TA, losartan, and TA-losartan combination downregulated the AT1R, Col IV, and TGF-ß1 expression and distribution in diabetic rat kidneys. Results suggest that TA is able to suppress diabetic nephropathy in rats effectively, probably by decreasing blood glucose levels and downregulating AT1R and TGF-ß1 expression.


Assuntos
Derivados de Alilbenzenos/farmacologia , Anisóis/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Nefropatias Diabéticas/tratamento farmacológico , Receptor Tipo 1 de Angiotensina/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Masculino , Ratos , Ratos Wistar
15.
Cell Cycle ; 20(3): 320-331, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33461373

RESUMO

Triple-negative breast cancer (TNBC) is the most aggressive histological subtype of breast cancer and is characterized by poor outcomes and a lack of specific-targeted therapies. Transforming growth factor-ß (TGF-ß) acts as the key cytokine in the epithelial-mesenchymal transition (EMT) and the metastasis of TNBC. However, the regulatory mechanisms of the TGF-ß signaling pathway remain largely unknown. In this study, we identified that the USP1/WDR48 complex could effectively enhance TGF-ß-mediated EMT and migration of TNBC cells. Furthermore, lower phosphorylation of Smad2/3, Erk, Jnk, and p38 was noted on the suppression of the expression of endogenous USP1 or WDR48. Moreover, the USP1-WDR48 complex was found to downregulate the polyubiquitination of TAK1 and mediate its in vitro stability. Therefore, our findings have shed a light on the novel role of the USP1/WDR48 complex in promoting TGF-ß-induced EMT and migration in TNBC via in vitro stabilization of TAK1.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , MAP Quinase Quinase Quinases/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Neoplasias de Mama Triplo Negativas/metabolismo , Proteases Específicas de Ubiquitina/biossíntese , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , MAP Quinase Quinase Quinases/genética , Estabilidade Proteica , Fator de Crescimento Transformador beta/genética , Neoplasias de Mama Triplo Negativas/genética , Proteases Específicas de Ubiquitina/genética
16.
Immunopharmacol Immunotoxicol ; 43(1): 85-93, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33406939

RESUMO

BACKGROUND: Previous studies have demonstrated that mifepristone in the daily low-dose affects the function of endometrium. These researches also implied an alteration of endometrium immune balance, which might be involved in regulating endometrial function. However, the detailed mechanisms remain to be further explored. METHODS: In this study, the expressions of CD80, CD86, and ICAM-1 in dendritic cells (DCs), which were stimulated with different concentrations of mifepristone (20, 65, and 200 nM), were detected by FACS. After that, we further evaluated the expression of Forkhead box P3 (FOXP3) and IL-10 in Tregs, which co-cultured with mifepristone treated DCs. In mechanism, we compared the indoleamine 2,3-dioxygenase (IDO) and TGF-ß expression with enzyme-linked immunosorbent assay (ELISA). RESULTS: The results indicated that mifepristone promoted the expressions of CD80, CD86, and ICAM-1 in a dosage dependent manner. Reversely, FOXP3 and IL-10 expression levels in Tregs co-cultured with mifepristone-treated DCs were significantly decreased compared with those co-cultured with nontreated DC. Furthermore, a significant reduce in IDO and TGF-ß expression was observed in DCs treated with mifepristone. By using the IDO inhibitor (1-methyl tryptophan, 1-MT) or TGF-b supplement, we confirmed that TGF-ß, but not IDO could rescue the downregulation of FOXP3 and IL-10 in Tregs co-cultured with mifepristone treated DCs. All of these results suggest that mifepristone may regulate DC function by decreasing TGF-ß expression, which further results in the downregulations of FOXP3 and IL-10 in Tregs. CONCLUSION: Therefore, our research provides a theoretical basis for a potentially clinical application of mifepristone as a novel contraceptive.


Assuntos
Células Dendríticas/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adulto , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Relação Dose-Resposta a Droga , Feminino , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/imunologia , Adulto Jovem
17.
Z Orthop Unfall ; 159(1): 54-66, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31746442

RESUMO

OBJECTIVE: TGFß1 plays an important role in the metabolism of articular cartilage and bone; however, the pathological mechanism and targets of TGFß1 in cartilage degradation and uncoupling of subchondral bone remodeling remain unclear. Therefore, in this study, we investigated the relationship between TGFß1 and major protein-degrading enzymes, and evaluated the role of high levels of active TGFß1 in the thickening of subchondral bone and calcification of articular cartilage. MATERIALS AND METHODS: The expression of TGFß1 and protein-degrading enzymes in clinical samples of articular cartilage and subchondral bone obtained from the knee joint of patients with osteoarthritis was detected by immunohistochemistry. The expression levels of TGFß1, MMP-3, MMP-13 and IL-1ß in cartilage and subchondral bone tissues were detected by absolute real-time quantitative RT-PCR. The expression of TGFß1, nestin and osterix in subchondral bone was detected by Western blot analysis and immunohistochemistry. The degree of subchondral bone thickening was determined by micro-computed tomography (CT) imaging. RESULTS: Expression of TGFß1 and cartilage-degrading enzymes was higher in the cartilage-disrupted group than that in the intact group. Furthermore, expression of TGFß1, nestin and osterix was significantly higher in the OA group than that in the control group. Micro-CT imaging showed that in the OA group, the subchondral bone plate is thickened and the density is increased. The trabecular bone structure is thick plate-like structure, the thickness of the trabecular bone is increased and the gap is small. CONCLUSIONS: The data suggest that highly active TGFß1 activates the expression of cartilage-degrading enzymes. Abnormally activated TGFß1 may induce formation of the subchondral bone and expansion of the calcified cartilage area, eventually leading to degradation of the cartilage tissue.


Assuntos
Ossos da Extremidade Inferior/metabolismo , Cartilagem Articular , Enzimas/metabolismo , Proteínas da Matriz Extracelular/biossíntese , Articulação do Joelho/metabolismo , Osteoartrite do Joelho , Fator de Crescimento Transformador beta/biossíntese , Ossos da Extremidade Inferior/diagnóstico por imagem , Calcinose/diagnóstico por imagem , Calcinose/metabolismo , Cartilagem Articular/diagnóstico por imagem , Cartilagem Articular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Imuno-Histoquímica , Articulação do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/metabolismo , Proteínas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Microtomografia por Raio-X
18.
Cell Biochem Funct ; 39(1): 116-125, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33006416

RESUMO

Uncoupling protein 1 (UCP1) is responsible for non-shivering thermogenesis, with restricted expression in brown/beige adipocytes in humans and rodents. We have previously shown an unexpected expression of UCP1 in bovine skeletal muscles. This study evaluated factors affecting Ucp1 gene expression in cultured bovine myogenic cells. Myosatellite cells, which were isolated from the bovine musculus longissimus cervicis, were induced to differentiate into myotubes in the presence of 2% horse serum. Previous studies using murine brown/beige adipocytes revealed that Ucp1 expression levels are directly increased by forskolin and all-trans retinoic acid (RA). The transforming growth factor-ß (TGF-ß)/activin pathway negatively regulated Ucp1 expression, whereas activation of the bone morphogenetic protein (BMP) pathway indirectly increases Ucp1 expression through the stimulation of brown/beige adipogenesis. Neither forskolin nor RA significantly affected Ucp1 mRNA levels in bovine myogenic cells. A-83-01, an inhibitor of the TGF-ß/activin pathway, stimulated myogenesis in these cells. A-83-01 significantly increased the expression of some brown fat signature genes such as Pgc-1α, Cox7a1, and Dio2, with a quantitative but not significant increase in the expression of Ucp1. Treatment with LDN-193189, an inhibitor of the BMP pathway, did not affect the differentiation of bovine myosatellite cells. Rather, LDN-193189 increased Ucp1 mRNA levels without modulating the levels of other brown/beige adipocyte-related genes. The current results indicate that the regulation of Ucp1 expression in bovine myogenic cells is distinct from that in murine brown/beige adipocytes, which has been more intensely characterized. SIGNIFICANCE OF THE STUDY: We previously reported unexpected expression of Ucp1 in bovine muscle tissues; Ucp1 expression has been known to be detected predominantly in brown/beige adipocytes. This study examined regulatory expression of bovine Ucp1 in myogenic cells. Consistent with the changes in expression levels of brown/beige adipocyte-selective genes, Ucp1 expression tended to be increased by inhibition of endogenous TGF-ß activity. In contrast, inhibition of endogenous BMP significantly increased Ucp1 expression without affecting brown/beige adipocyte-selective gene expression. The current results indicate that regulatory expression of Ucp1 in bovine myogenic cells is distinct from that in murine brown/beige adipocytes that is more intensely characterized.


Assuntos
Regulação da Expressão Gênica , Mioblastos Esqueléticos/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Proteína Desacopladora 1/biossíntese , Animais , Bovinos , Células Cultivadas , Mioblastos Esqueléticos/citologia
19.
J Chin Med Assoc ; 84(1): 51-60, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33177403

RESUMO

BACKGROUND: Liposomes containing docosahexaenoic acid (DHA) and phosphatidylserine were claimed to inhibit osteoclast formation and bone resorption in the inflammatory status. Herein, we proposed that an apoptotic mimicry (SQ liposome) prepared from squid-skin phospholipids can explore the suppressive osteoclastogenesis. METHODS: The intermolecular fatty-acid composition in the phospholipid of squid-skin extract was analyzed by GC-FID. The SQ liposome structure was characterized by size distribution and zeta potential (ζ). RAW 264.7 cell is used to study the effect of SQ liposomes on osteoclast differentiation. Secretion of prostaglandin E2 (PGE2) and transforming growth factor-ß (TGF-ß) from RAW 264.7 cells were assayed. Antiosteoclastogenesis effects were performed via the tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cell (MNC) counting, bone resorption pit assay, and TRAP activity analysis. The specific gene expressions related to antiosteoclastogenesis were also detected. RESULTS: An apoptotic mimicry through the use of a single-layer liposome (SQ liposome) with phosphatidylserine exposure contains DHA (28.7%) and eicosapentaenoic acid (EPA, 11.8%). Co-treatment with receptor activator of nuclear factor kappa B ligand (RANKL)/macrophage colony-stimulating factor induced RAW 264.7-cell differentiation into mature osteoclasts, thus enhancing PGE2 and TGF-ß secretion. However, cotreatment with 1 mg/mL of SQ liposome restored (p < 0.05) the cell viabilities under the RANKL stress. Increased PGE2 levels was downregulated (p < 0.05) in cotreatments with 0.11 and 0.33 mg/mL of SQ liposome, but on the TGF-ß levels were not (p > 0.05) influenced in SQ liposome cotreatments. Cotreatments with 0.33-1 mg/mL of SQ liposome suppressed (p < 0.05) the osteoclast maturation (such as decreased MNCs and bone pit formation), inhibited TRAP activities, and downregulated the osteoclastogenesis-related gene expressions. CONCLUSION: In summary, current data support that a possible prevention of our prepared SQ liposomes which are rich in DHA and EPA on bone loss is through the suppression of osteoclastogenesis. Moreover, based on the results from this study an in vivo study warrants a further investigation.


Assuntos
NF-kappa B/fisiologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fosfolipídeos/farmacologia , Animais , Decapodiformes/metabolismo , Dinoprostona/biossíntese , Lipossomos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Osteoclastos/fisiologia , Ligante RANK/farmacologia , Células RAW 264.7 , Pele/metabolismo , Fosfatase Ácida Resistente a Tartarato/metabolismo , Fator de Crescimento Transformador beta/biossíntese
20.
Int Arch Allergy Immunol ; 182(4): 277-291, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33147596

RESUMO

INTRODUCTION: The use of probiotics has been broadly popularized due to positive effects in the attenuation of aberrant immune responses such as asthma. Allergic asthma is a chronic respiratory disease characterized by airway inflammation and remodelling. OBJECTIVE: This study was aimed to evaluate the effect of oral administration of Lactococcus lactis NZ9000 on asthmatic airway inflammation and lung tissue remodelling in rats and its relation to the maintenance of an adequate intestinal barrier. METHODS: Wistar rats were ovalbumin (OVA) sensitized and challenged and orally treated with L. lactis. Lung inflammatory infiltrates and cytokines were measured, and remodelling was evaluated. Serum OVA-specific immunoglobulin (Ig) E levels were assessed. We also evaluated changes on intestinal environment and on systemic immune response. RESULTS: L. lactis diminished the infiltration of proinflammatory leucocytes, mainly eosinophils, in the bronchoalveolar compartment, decreased lung IL-4 and IL-5 expression, and reduced the level of serum allergen-specific IgE. Furthermore, L. lactis prevented eosinophil influx, collagen deposition, and goblet cell hyperplasia in lung tissue. In the intestine, L. lactis-treated asthmatic rats increased Peyer's patch and goblet cell quantity and mRNA expression of IgA, MUC-2, and claudin. Additionally, intestinal morphological alterations were normalized by L. lactis administration. Splenocyte proliferative response to OVA was abolished, and serum levels of transforming growth factor (TGF)-ß were increased by L. lactis treatment. CONCLUSIONS: These findings suggest that L. lactis is a potential candidate for asthma prevention, and the effect is mediated by the improvement of intestinal barrier function and systemic TGF-ß production.


Assuntos
Remodelação das Vias Aéreas , Asma/metabolismo , Asma/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lactococcus lactis/fisiologia , Probióticos/administração & dosagem , Fator de Crescimento Transformador beta/biossíntese , Remodelação das Vias Aéreas/imunologia , Animais , Asma/etiologia , Asma/prevenção & controle , Citocinas/metabolismo , Modelos Animais de Doenças , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Mediadores da Inflamação/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Ovalbumina/imunologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...