Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Int J Mol Sci ; 25(5)2024 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-38474186

RESUMO

Programmed death ligand 1 (PD-L1) plays a pivotal role in cancer immune evasion and is a critical target for cancer immunotherapy. This review focuses on the regulation of PD-L1 through the dynamic processes of ubiquitination and deubiquitination, which are crucial for its stability and function. Here, we explored the intricate mechanisms involving various E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) that modulate PD-L1 expression in cancer cells. Specific ligases are discussed in detail, highlighting their roles in tagging PD-L1 for degradation. Furthermore, we discuss the actions of DUBs that stabilize PD-L1 by removing ubiquitin chains. The interplay of these enzymes not only dictates PD-L1 levels but also influences cancer progression and patient response to immunotherapies. Furthermore, we discuss the therapeutic implications of targeting these regulatory pathways and propose novel strategies to enhance the efficacy of PD-L1/PD-1-based therapies. Our review underscores the complexity of PD-L1 regulation and its significant impact on the tumor microenvironment and immunotherapy outcomes.


Assuntos
Antígeno B7-H1 , Neoplasias , Humanos , Imunoterapia , Ubiquitinação , Ubiquitina-Proteína Ligases , Ubiquitina , Microambiente Tumoral
2.
Int J Mol Sci ; 24(2)2023 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-36674651

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease of the joint synovial membranes. RA is difficult to prevent or treat; however, blocking proinflammatory cytokines is a general therapeutic strategy. Pulsed electromagnetic field (PEMF) is reported to alleviate RA's inflammatory response and is being studied as a non-invasive physical therapy. In this current study, PEMF decreased paw inflammation in a collagen-induced arthritis (CIA) murine model. PEMF treatment at 10 Hz was more effective in ameliorating arthritis than at 75 Hz. In the PEMF-treated CIA group, the gross inflammation score and cartilage destruction were lower than in the untreated CIA group. The CIA group treated with PEMF also showed lower serum levels of IL-1ß but not IL-6, IL-17, or TNF-α. Serum levels of total anti-type II collagen IgG and IgG subclasses (IgG1, IgG2a, and IgG2b) remained unchanged. In contrast, tissue protein levels of IL-1ß, IL-6, TNF-α, receptor activator of nuclear factor kappa-Β (RANK), RANK ligand (RANKL), IL-6 receptor (IL-6R), and TNF-α receptor1 (TNFR1) were all lower in the ankle joints of the PEMF-treated CIA group compared with the CIA group. The results of this study suggest that PEMF treatment can preserve joint morphology cartilage and delay the occurrence of CIA. PEMF has potential as an effective adjuvant therapy that can suppress the progression of RA.


Assuntos
Artrite Experimental , Artrite Reumatoide , Camundongos , Animais , Artrite Experimental/tratamento farmacológico , Fator de Necrose Tumoral alfa/uso terapêutico , Modelos Animais de Doenças , Campos Eletromagnéticos , Citocinas , Artrite Reumatoide/tratamento farmacológico , Inflamação/tratamento farmacológico , Imunoglobulina G/uso terapêutico
3.
Int J Mol Sci ; 25(1)2023 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-38203534

RESUMO

Enterotoxigenic Bacteroides fragilis (ETBF) causes colitis and is implicated in inflammatory bowel diseases and colorectal cancer. The ETBF-secreted B. fragilis toxin (BFT) causes cleavage of the adherence junction, the E-cadherin, resulting in the large intestine showing IL-17A inflammation in wild-type (WT) mice. However, intestinal pathology by ETBF infection is not fully understood in B-cell-deficient mice. In this study, ETBF-mediated inflammation was characterized in B-cell-deficient mice (muMT). WT or muMT C57BL/6J mice were orally inoculated with ETBF and examined for intestinal inflammation. The indirect indicators for colitis (loss of body weight and cecum weight, as well as mortality) were increased in muMT mice compared to WT mice. Histopathology and inflammatory genes (Nos2, Il-1ß, Tnf-α, and Cxcl1) were elevated and persisted in the large intestine of muMT mice compared with WT mice during chronic ETBF infection. However, intestinal IL-17A expression was comparable between WT and muMT mice during infection. Consistently, flow cytometry analysis applied to the mesenteric lymph nodes showed a similar Th17 immune response in both WT and muMT mice. Despite elevated ETBF colonization, the ETBF-infected muMT mice showed no histopathology or inflammation in the small intestine. In conclusion, B cells play a protective role in ETBF-induced colitis, and IL-17A inflammation is not attributed to prompted colitis in B-cell-deficient mice. Our data support the fact that B cells are required to ameliorate ETBF infection-induced colitis in the host.


Assuntos
Infecções Bacterianas , Colite , Animais , Camundongos , Camundongos Endogâmicos C57BL , Bacteroides fragilis , Interleucina-17/genética , Inflamação
4.
Biomater Sci ; 10(15): 4335-4344, 2022 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-35775589

RESUMO

Despite advances in cancer therapy, the discovery of effective cancer treatments remains challenging. In this study, a simple method was developed to increase the efficiency of doxorubicin (DOX) delivery in a lung metastasis model. This method comprises a simple configuration to increase the delivery efficiency via precise engineering of the size, shape, loading content, and biodegradability of the drug delivery system. This system had a 3 µm discoidal shape and exerted approximately 90% burst release of the drug within the first 24 h. There was no cytotoxicity of the drug carrier up to a concentration of 1 mg ml-1, and DOX from the carrier was delivered into the cancer cells, exhibiting an anticancer effect comparable to that of the free drug. The ex vivo results revealed a strong correlation between the location of cancer cells in the lung and the location of DOX delivered by this drug delivery system. These drug carriers were confirmed to intensively deliver DOX to cancer cells in the lung, with minimal off-target effects. These findings indicate that this delivery system can be a new approach to improving the survival rate and reducing the side effects caused by anticancer drugs without the use of targeting ligands and polyethylene glycol.


Assuntos
Doxorrubicina , Neoplasias Pulmonares , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Portadores de Fármacos , Sistemas de Liberação de Medicamentos/métodos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Polietilenoglicóis/farmacologia , Polímeros , Taxa de Sobrevida
5.
Biomedicines ; 10(4)2022 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-35453577

RESUMO

Enterotoxigenic Bacteroides fragilis (ETBF) has emerged as a gut microbiome pathogen that can promote colitis associated cancer in humans. ETBF secretes the metalloprotease, B. fragilis toxin (BFT), which can induce ectodomain cleavage of E-cadherin and IL-8 secretion through the ß-catenin, NF-κB, and MAPK pathways in intestinal epithelial cells. However, it is still unclear whether E-cadherin cleavage is required for BFT induced IL-8 secretion and the relative contribution of these signaling pathways to IL-8 secretion. Using siRNA knockdown and CRISPR knockout studies, we found that E-cadherin cleavage is required for BFT mediated IL-8 secretion. In addition, genetic ablation of ß-catenin indicates that ß-catenin is required for the BFT induced increase in transcriptional activity of NF-κB, p65 nuclear localization and early IL-8 secretion. These results suggest that BFT induced ß-catenin signaling is upstream of NF-κB activation. However, despite ß-catenin gene disruption, BFT still activated the MAPK pathway, suggesting that the BFT induced activation of the MAPK signaling pathway is independent from the E-cadherin/ß-catenin/NF-κB pathway. These findings show that E-cadherin and ß-catenin play a critical role in acute inflammation following ETBF infection through the inflammatory response to BFT in intestinal epithelial cells.

6.
Int J Med Sci ; 19(2): 353-363, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35165521

RESUMO

Cultured human skeletal-muscle satellite cells have properties of mesenchymal stem cells (skeletal muscle satellite cell-derived mesenchymal stem cells, SkMSCs) and play anti-inflammatory roles by secreting prostaglandin E2 and hepatocyte growth factor (HGF). To evaluate the utility of SkMSCs in treating liver diseases, we determined whether SkMSCs could ameliorate acute liver and gut inflammation induced by binge ethanol administration. Binge drinking of ethanol led to weight loss in the body and spleen, liver inflammation and steatosis, and increased serum ALT and AST levels (markers of liver injury), along with increased IL-1ß, TNF-α, and iNOS expression levels in mice. However, levels of these binge-drinking-induced indicators were reduced by a single intraperitoneal treatment of SkMSCs. Furthermore, levels of bacteria-derived lipopolysaccharide decreased in the livers and sera of ethanol-exposed mice after SkMSC administration. SkMSCs decreased the extent of tissue inflammation and reduced villus and crypt lengths in the small intestine after alcohol binge drinking. SkMSCs also reduced the leakage of blood albumin, an indicator of leaky gut, in the stool of ethanol-exposed mice. Alcohol-induced damage to human colonic Caco-2/tc7 cells was also alleviated by HGF. Therefore, a single treatment with SkMSCs can attenuate alcoholic liver damage by reducing inflammatory responses in the liver and gut, suggesting that SkMSCs could be used in cell therapy to treat alcoholic liver diseases.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/sangue , Etanol/efeitos adversos , Hepatopatias Alcoólicas/terapia , Transplante de Células-Tronco Mesenquimais , Células Satélites de Músculo Esquelético/transplante , Animais , Consumo Excessivo de Bebidas Alcoólicas/complicações , Células CACO-2 , Células Cultivadas , Dinoprostona/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Inflamação , Fígado/metabolismo , Hepatopatias Alcoólicas/etiologia , Células-Tronco Mesenquimais , Camundongos
7.
Cell Biol Int ; 45(12): 2443-2451, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34374483

RESUMO

Skeletal muscle satellite cells (SkMSCs) play crucial roles in muscle fiber maintenance, repair, and remodeling; however, it remains unknown if these properties are preserved in cultured SkMSCs. In this study, we investigated the characteristics of cultured SkMSCs and their ability to regulate the activity of M1 macrophages. SkMSCs grew well with an average population doubling time of 26.26 ± 6.85 h during 10 passages (P). At P5, Pax7, MyoD, cluster of differentiation (CD)34, and CD56 were not expressed in SkMSCs, but the MSC markers CD73, CD105, and CD90 were expressed and the cells were differentiated into adipocytes and osteoblasts. When SkMSCs were cocultured with macrophages, interleukin (IL)-1ß secretion was decreased, prostaglandin (PG)E2 was produced in coculture, and cyclooxygenase-2 protein was induced in an SkMSC-dependent manner. Hepatocyte growth factor (HGF) was highly secreted by monocultured SkMSCs; interferon-γ and lipopolysaccharide reduced its expression level. However, HGF expression recovered when SkMSCs and macrophages were cocultured. Although exogenous PGE2 upregulated macrophage pro-IL-1ß expression, it suppressed the secretion of cleaved IL-1ß. In contrast, HGF decreased active IL-1ß secretion without affecting pro-IL-1ß expression. Co-treatment of macrophages with HGF and PGE2 reduced pro-IL-1ß expression level and active IL-1ß secretion. Our results suggest that SkMSCs lose their satellite cell properties during serial passaging but acquire mesenchymal stem cell properties including the ability to exert an anti-inflammatory response for macrophages through PGE2 and HGF.


Assuntos
Anti-Inflamatórios/metabolismo , Dinoprostona/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Tecido Adiposo/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Hepatócitos/metabolismo , Humanos , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Células THP-1/metabolismo
8.
Neoplasia ; 23(6): 561-573, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34077834

RESUMO

Previously, we found that rosmarinic acid (RA) exerted anti-inflammatory activities in a dextran sulfate sodium (DSS)-induced colitis model. Here, we investigated the anti-tumor effects of RA on colitis-associated colon cancer (CAC) and the underlying molecular mechanisms. We established an azoxymethane (AOM)/DSS-induced CAC murine model for in vivo studies and used a conditioned media (CM) culture system in vitro. H&E staining, immunohistochemistry, western blot assay, enzyme-linked immunosorbent assay, molecular docking, co-immunoprecipitation, and immunofluorescence assay were utilized to investigate how RA prevented colorectal cancer. In the AOM/DSS-induced CAC murine model, RA significantly reduced colitis severity, inflammation-related protein expression, tumor incidence, and colorectal adenoma development. It significantly modulated toll-like receptor-4 (TLR4)-mediated nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) activation, thus attenuating the expression of anti-apoptotic factors, which mediate transcription factor-dependent tumor growth. In vitro, RA inhibited CM-induced TLR4 overexpression and competitively inhibited TLR4-myeloid differentiation factor 2 complex in an inflammatory microenvironment. Thus, RA suppressed NF-κB and STAT3 activation in colon cancer cells in an inflammatory microenvironment. Therefore, RA suppressed colitis-associated tumorigenesis in the AOM/DSS-induced CAC murine model and abrogated human colon cancer progression in an inflammatory microenvironment by propitiating TLR4-mediated NF-κB and STAT3 activation, pleiotropically.


Assuntos
Cinamatos/farmacologia , Neoplasias Associadas a Colite/etiologia , Neoplasias Associadas a Colite/metabolismo , Depsídeos/farmacologia , NF-kappa B/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Animais , Biomarcadores , Linhagem Celular Tumoral , Cinamatos/química , Neoplasias Associadas a Colite/tratamento farmacológico , Neoplasias Associadas a Colite/patologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/etiologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Depsídeos/química , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Modelos Moleculares , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto , Ácido Rosmarínico
9.
Anticancer Res ; 41(6): 2859-2866, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34083276

RESUMO

BACKGROUND/AIM: Genetic manipulation of stem cells using non-viral vectors is still limited due to low transfection efficiency. We investigated whether the DNA-binding cell-permeation peptides (CPP) can enhance the transfection efficiency of non-viral vectors in adipose tissue-derived mesenchymal stem cells (ASCs) and whether ASCs over-expressing TRAIL through CPP can inhibit the growth of glioma U251MG cells in vitro and in vivo. MATERIALS AND METHODS: ASCs were genetically engineered to over-express TRAIL by using CPP, pCMV3-TRAIL and lipid-based transfection reagents (X-tremeGENE). RESULTS: The transfection efficiency of ASCs increased by approximately 7% using CPP; 53.9% of ASCs were transfected and TRAIL expression in ASCs increased by approximately 3 times compared to X-tremeGENE alone. ASCs over-expressing TRAIL using CPP inhibited growth of glioma U251MG cells both in vitro and in the U251MG xenograft model. CONCLUSION: CPP can be used as an enhancer for genetically manipulating ASCs and tumor treatment.


Assuntos
Tecido Adiposo/citologia , Neoplasias Encefálicas/patologia , Peptídeos Penetradores de Células/metabolismo , DNA/metabolismo , Glioma/patologia , Células-Tronco Mesenquimais/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Genomics Proteomics ; 18(4): 569-578, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34183389

RESUMO

BACKGROUND/AIM: Mesenchymal stem cell-based tumor therapy is still limited due to the insufficient secretion of effectors and discrepancies between their in vitro and in vivo efficacy. We investigated whether genetically engineered adipose tissue-derived mesenchymal stem cells (ASCs) overexpressing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) had inhibitory effects on H460 tumor growth both in vitro and in an H460 xenograft model. MATERIALS AND METHODS: Genetically engineered adipose tissue-derived mesenchymal stem cells (ASCs) overexpressing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) were obtained from plasmid transfection with pCMV3-TRAIL and -interferon (IFN)-ß (producing ASC-TRAIL and ASC-IFN-ß, respectively). Death of H460 cells co-cultured with ASCs, ASC-TRAIL, and ASC-IFN-ß or exposed to their conditioned medium was evaluated via apoptosis and cytotoxicity assays. In addition, in an H460 xenograft model (n=10 per group), the antitumor potential of TRAIL-overexpressing, and IFN-ß-overexpressing ASCs was investigated. RESULTS: Conditioned medium obtained from ASC-IFN-ß increased apoptosis of H460 cells more than did ASC-TRAIL. Additionally, in H460 xenograft models, while native ASCs promoted tumor growth, ASC-TRAIL and ASC-IFN-ß both dramatically suppressed tumor growth. Interestingly, in the context of ASC-IFN-ß, tumors were detected only in 20% of nude mice, with smaller sizes and lower weights than those of the control group. CONCLUSION: TRAIL-overexpressing ASCs can be used to treat tumors; ASC-IFN-ß in particular secrete a higher level of TRAIL.


Assuntos
Tecido Adiposo/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Tecido Adiposo/citologia , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/metabolismo , Meios de Cultivo Condicionados/farmacologia , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Camundongos , Camundongos Nus , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Pharmaceutics ; 12(12)2020 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-33327415

RESUMO

A layered double hydroxide (LDH)-based anticancer delivery system was investigated in terms of crystalline phase, particle size, hydrodynamic radius, zeta potential, etc. through in vitro and in vivo study. Size controlled LDH with anticancer drug methotrexate (MTX) incorporation was successfully prepared through step-by-step hydrothermal reaction and ion-exchange reaction. The MTX-LDH was determined to have a neutral surface charge and strong agglomeration in the neutral aqueous condition due to the surface adsorbed MTX; however, the existence of proteins in the media dramatically reduced agglomeration, resulting in the hydrodynamic radius of MTX-LDH being similar to the primary particle size. The protein fluorescence quenching assay exhibited that MTX readily reduced the fluorescence of proteins, suggesting that the interaction between MTX and proteins was strong. On the other hand, MTX-LDH showed much less binding constant to proteins compared with MTX, implying that the protein interaction of MTX was effectively blocked by the LDH carrier. The in vivo hemolysis assay after intravenous injection of MTX-LDH showed neither significant reduction in red blood cell number nor membrane damage. Furthermore, the morphology of red blood cells in a mouse model did not change upon MTX-LDH injection. Scanning electron microscopy showed that the MTX-LDH particles were attached on the blood cells without serious denaturation of cellular morphology, taking advantage of the cell hitchhiking property.

12.
Sci Rep ; 10(1): 20321, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33230134

RESUMO

We report a technique to reconstruct cardiovascular tissue using multiscale scaffolds incorporating polycaprolactone fibers with double-layered hydrogels comprising fibrin hydrogel surrounded by secondary alginate hydrogel. The scaffolds compartmentalized cells into the core region of cardiac tissue and the peripheral region of blood vessels to construct cardiovascular tissue, which was accomplished by a triple culture system of adipose-derived mesenchymal stem cells (ADSCs) with C2C12 myoblasts on polycaprolactone (PCL) fibers along with human umbilical vein endothelial cells (HUVECs) in fibrin hydrogel. The secondary alginate hydrogel prevented encapsulated cells from migrating outside scaffold and maintained the scaffold structure without distortion after subcutaneous implantation. According to in vitro studies, resultant scaffolds promoted new blood vessel formation as well as cardiomyogenic phenotype expression of ADSCs. Cardiac muscle-specific genes were expressed from stem cells and peripheral blood vessels from HUVECs were also successfully developed in subcutaneously implanted cell-laden multiscale scaffolds. Furthermore, the encapsulated stem cells modulated the immune response of scaffolds by secreting anti-inflammatory cytokines for successful tissue construction. Our study reveals that multiscale scaffolds can be promising for the remodeling and transplantation of cardiovascular tissue.


Assuntos
Fibrina/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Hidrogéis/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mioblastos/metabolismo , Poliésteres/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Doenças Cardiovasculares/terapia , Linhagem Celular , Citocinas/metabolismo , Humanos , Células-Tronco Mesenquimais/imunologia , Camundongos , Neovascularização Fisiológica , Fenótipo , Transplante de Tecidos/métodos
13.
Int J Mol Sci ; 21(21)2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33126615

RESUMO

Consumption of a Western-type diet has been linked to gut-microbiota-mediated colon inflammation that constitutes a risk factor for colorectal cancer. A high salt diet (HSD) exacerbates IL-17A-induced inflammation in inflammatory bowel disease and other autoimmune diseases. Enterotoxigenic Bacteroides fragilis (ETBF) is a gut commensal bacterium and reported to be a potent initiator of colitis via secretion of the Bacteroides fragilis toxin (BFT). BFT induces ectodomain cleavage of E-cadherin in colonic epithelial cells, consequently leading to cell rounding, epithelial barrier disruption, and the secretion of IL-8, which promotes tumorigenesis in mice via IL-17A-mediated inflammation. A HSD is characteristic of the Western-type diet and can exhibit inflammatory effects. However, a HSD induces effects in ETBF-induced colitis and tumorigenesis remain unknown. In this study, we investigated HSD effects in ETBF-colonized mice with azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced tumorigenesis as well as ETBF colitis mice. Unexpectedly, ETBF-infected mice fed a HSD exhibited decreased weight loss and splenomegaly and reduction of colon inflammation. The HSD significantly decreased the expression of IL-17A and inducible nitric oxide synthase (iNOS) in the colonic tissues of ETBF-infected mice. In addition, serum levels of IL-17A and nitric oxide (NO) were also diminished. However, HT29/C1 colonic epithelial cells treated with sodium chloride showed no changes in BFT-induced cellular rounding and IL-8 expression. Furthermore, HSD did not affect ETBF colonization in mice. In conclusion, HSD decreased ETBF-induced tumorigenesis through suppression of IL-17A and iNOS expression in the colon. HSD also inhibited colonic polyp numbers in the ETBF-infected AOM/DSS mice. Taken together, these findings suggest that a HSD consumption inhibited ETBF-promoted colon carcinogenesis in mice, indicating that a HSD could have beneficial effects under certain conditions.


Assuntos
Infecções por Bacteroides/complicações , Bacteroides fragilis/patogenicidade , Carcinogênese/imunologia , Neoplasias do Colo/prevenção & controle , Inflamação/prevenção & controle , Cloreto de Sódio na Dieta/administração & dosagem , Animais , Infecções por Bacteroides/microbiologia , Carcinogênese/patologia , Neoplasias do Colo/etiologia , Neoplasias do Colo/patologia , Feminino , Inflamação/etiologia , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL
14.
Int J Mol Sci ; 21(11)2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32485960

RESUMO

We have previously reported that adipose tissue-derived stem cells (ASCs) cultured at high cell density can induce cancer cell death through the expression of type I interferons and tumor necrosis factor (TNF)-related apoptosis-inducing ligands (TRAIL). Here, we investigated whether TRAIL-expressing ASCs induced by M1 macrophages can alleviate colitis-associated cancer in an azoxymethane (AOM)/dextran sodium sulfate (DSS) animal model. M1 macrophages significantly increased the TRAIL expression in ASCs, which induced the apoptosis of LoVo cells in a TRAIL-dependent manner. However, CD133knockout LoVo cells, generated using the CRISPR-Cas9 gene-editing system, were resistant to TRAIL. In the AOM/DSS-induced colitis-associated cancer model, the intraperitoneal transplantation of TRAIL-expressing ASCs significantly suppressed colon cancer development. Moreover, immunohistochemical staining revealed a low CD133 expression in tumors from the AOM/DSS + ASCs group when compared with tumors from the untreated group. Additionally, the ASC treatment selectively reduced the number of M2 macrophages in tumoral (45.7 ± 4.2) and non-tumoral mucosa (30.3 ± 1.5) in AOM/DSS + ASCs-treated animals relative to those in the untreated group (tumor 71.7 ± 11.2, non-tumor 94.3 ± 12.5; p < 0.001). Thus, TRAIL-expressing ASCs are promising agents for anti-tumor therapy, particularly to alleviate colon cancer by inducing the apoptosis of CD133+ cancer stem cells and decreasing the M2 macrophage population.


Assuntos
Apoptose , Neoplasias Associadas a Colite/metabolismo , Colite/complicações , Macrófagos/metabolismo , Células-Tronco/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Antígeno AC133/metabolismo , Tecido Adiposo/citologia , Adulto , Animais , Azoximetano , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Colite/metabolismo , Neoplasias Associadas a Colite/complicações , Colo/patologia , Sulfato de Dextrana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neoplásicas/citologia
15.
Int J Med Sci ; 17(5): 609-619, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32210710

RESUMO

Interferon (IFN)-ß and/or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) secreted by adipose tissue-derived mesenchymal stem cells (ASCs) have been proposed as key mechanistic factors in anti-cancer efficacy in lung cancer and breast cancer cells, where they act through paracrine signaling. We hypothesized that IFN-ß and TRAIL produced by ASCs suppress proliferation of hepatocellular carcinoma cells (HCCs). The present study evaluated the anti-cancer effects of ASCs on HCCs in vitro. We found that indirect co-culture with ASCs diminished growth of Huh7 hepatocellular carcinoma cells with increased protein levels of p53/p21 and phosphorylated STAT1 (pSTAT1), without apoptosis. Treatment with ASC-conditioned medium (ASC-CM) also decreased growth of Huh7 cells through elevated p53/p21 and pSTAT1 signaling. ASC-CM-mediated inhibition of cell growth was neutralized in Huh7 cells treated with anti-IFN-ß antibody compared to that in ASC-CM-treated Huh7 cells incubated with an anti-TRAIL antibody. Treatment with JAK1/JAK2 inhibitors recovered inhibition of growth in Huh7 cells incubated in ASC-CM or IFN-ß via down-regulation of pSTAT1/p53/p21. However, treatment of IFN-ß resulted in no alterations in resistance of Huh7 cells to TRAIL. Our findings suggest that ASCs decrease growth through activated STAT1-mediated p53/p21 by IFN-ß, but not TRAIL, in Huh7 cells.


Assuntos
Carcinoma Hepatocelular/terapia , Interferon beta/metabolismo , Neoplasias Hepáticas/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Humanos , Janus Quinases/metabolismo , Neoplasias Hepáticas/metabolismo , Células-Tronco Mesenquimais/citologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
16.
Int J Med Sci ; 17(2): 145-152, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32038097

RESUMO

The azoxymethane (AOM)/dextran sulfate sodium (DSS) murine model is commonly used to study colitis-associated cancer. The human commensal bacterium, enterotoxigenic Bacteroides fragilis (ETBF) secretes the Bacteroides fragilis toxin (BFT) which is necessary and sufficient to cause colitis. We report that BALB/c mice infected with WT-ETBF and administered three cycles of AOM/DSS developed numerous, large-sized polyps predominantly in the colorectal region. In addition, AOM/DSS-treated BALB/c mice orally inoculated with wild-type nontoxigenic Bacteroides fragilis (WT-NTBF) overexpressing bft (rETBF) developed numerous polyps whereas mice infected with WT-NTBF overexpressing a biologically inactive bft (rNTBF) did not promote polyp formation. Unexpectedly, the combination of AOM+ETBF did not induce polyp formation whereas ETBF+DSS did induce polyp development in a subset of BALB/c mice. In conclusion, WT-ETBF promoted polyp development in AOM/DSS murine model with increased colitis in BALB/c mice. The model described herein provides an experimental platform for understanding ETBF-induced colonic tumorigenesis and studying colorectal cancer in wild-type mice.


Assuntos
Infecções por Bacteroides/patologia , Carcinogênese/genética , Colite/patologia , Neoplasias Colorretais/patologia , Animais , Azoximetano/toxicidade , Toxinas Bacterianas/toxicidade , Infecções por Bacteroides/induzido quimicamente , Infecções por Bacteroides/complicações , Infecções por Bacteroides/microbiologia , Bacteroides fragilis/patogenicidade , Carcinogênese/induzido quimicamente , Colite/induzido quimicamente , Colite/complicações , Colite/microbiologia , Colo/efeitos dos fármacos , Colo/patologia , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/complicações , Neoplasias Colorretais/microbiologia , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Humanos , Metaloendopeptidases/toxicidade , Camundongos , Pólipos/induzido quimicamente
17.
Int J Mol Sci ; 21(3)2020 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-32013191

RESUMO

Chronic inflammation has been linked to colitis-associated colorectal cancer in humans. The human symbiont enterotoxigenic Bacteroides fragilis (ETBF), a pro-carcinogenic bacterium, has the potential to initiate and/or promote colorectal cancer. Antibiotic treatment of ETBF has shown promise in decreasing colonic polyp formation in murine models of colon cancer. However, there are no reported natural products that have shown efficacy in decreasing polyp burden. In this study, we investigated the chemopreventive effects of oral administration of zerumbone in ETBF-colonized mice with azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced tumorigenesis. Zerumbone significantly reduced the severity of disease activity index (DAI) scores as well as several parameters of colonic inflammation (i.e., colon weight, colon length, cecum weight and spleen weight). In addition, inflammation of the colon and cecum as well as hyperplasia was reduced. Zerumbone treatment significantly inhibited colonic polyp numbers and prevented macroadenoma progression. Taken together, these findings suggest that oral treatment with zerumbone inhibited ETBF-promoted colon carcinogenesis in mice indicating that zerumbone could be employed as a promising protective agent against ETBF-mediated colorectal cancer.


Assuntos
Bacteroides fragilis/patogenicidade , Neoplasias do Colo/prevenção & controle , Substâncias Protetoras/uso terapêutico , Sesquiterpenos/uso terapêutico , Administração Oral , Animais , Azoximetano/toxicidade , Peso Corporal/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Colite/complicações , Colite/microbiologia , Colite/patologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Sulfato de Dextrana/toxicidade , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Substâncias Protetoras/farmacologia , Sesquiterpenos/farmacologia , Índice de Gravidade de Doença
18.
Int J Mol Sci ; 20(18)2019 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-31540059

RESUMO

Enterotoxigenic Bacteroides fragilis (ETBF) is human intestinal commensal bacterium and a potent initiator of colitis through secretion of the metalloprotease Bacteroides fragilis toxin (BFT). BFT induces cleavage of E-cadherin in colon cells, which subsequently leads to NF-κB activation. Zerumbone is a key component of the Zingiber zerumbet (L.) Smith plant and can exhibit anti-bacterial and anti-inflammatory effects. However, whether zerumbone has anti-inflammatory effects in ETBF-induced colitis remains unknown. The aim of this study was to determine the anti-inflammatory effect of orally administered zerumbone in a murine model of ETBF infection. Wild-type C57BL/6 mice were infected with ETBF and orally administered zerumbone (30 or 60 mg/kg) once a day for 7 days. Treatment of ETBF-infected mice with zerumbone prevented weight loss and splenomegaly and reduced colonic inflammation with decreased macrophage infiltration. Zerumbone treatment significantly decreased expression of IL-17A, TNF-α, KC, and inducible nitric oxide synthase (iNOS) in colonic tissues of ETBF-infected mice. In addition, serum levels of KC and nitrite was also diminished. Zerumbone-treated ETBF-infected mice also showed decreased NF-κB signaling in the colon. HT29/C1 colonic epithelial cells treated with zerumbone suppressed BFT-induced NF-κB signaling and IL-8 secretion. However, BFT-mediated E-cadherin cleavage was unaffected. Furthermore, zerumbone did not affect ETBF colonization in mice. In conclusion, zerumbone decreased ETBF-induced colitis through inhibition of NF-κB signaling.


Assuntos
Antibacterianos/uso terapêutico , Infecções por Bacteroides/tratamento farmacológico , Bacteroides fragilis , Colite/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Sesquiterpenos/uso terapêutico , Animais , Toxinas Bacterianas , Infecções por Bacteroides/imunologia , Bacteroides fragilis/metabolismo , Caderinas/metabolismo , Colite/imunologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/fisiopatologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Células HT29 , Humanos , Interleucina-17/metabolismo , Interleucina-8/sangue , Metaloendopeptidases , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
19.
Cancer Lett ; 440-441: 202-210, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30393160

RESUMO

Although mesenchymal stem cells (MSCs) have been reported to inhibit tumor growth, the mechanism controlling this tumor suppression function is unclear. Here, we report that high-density (40,000 cells/cm2) cultured adipose tissue-derived MSCs (40K-ASCs) expressed interferon (IFN)-ß and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL); we also found that serum deprivation during cell culture induced the expression of IFN-ß and TRAIL. In addition, the mRNA expression of IFN-ß, but not TRAIL, was increased during the washing step required for the transplantation of normal-density (5000 cells/cm2) cultured ASCs (5K-ASCs). When the human lung cancer cell line H460 was co-cultured with 40K-ASCs, necrotic cell death was dramatically increased in vitro. When ASCs were injected after four washes, both 5K-ASCs and 40K-ASCs substantially reduced tumor weight in H460-derived cancer animal models. These results suggest that serum deprivation during the culture of 40K-ASCs or during the washing step of 5K-ASCs can induce IFN-ß and/or TRAIL expression, ultimately leading to the tumor suppression capability of ASCs.


Assuntos
Interferon beta/biossíntese , Neoplasias Pulmonares/terapia , Células-Tronco Mesenquimais/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Adulto , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Interferon beta/genética , Interferon beta/farmacologia , Neoplasias Pulmonares/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Adulto Jovem
20.
Biochem Biophys Res Commun ; 498(4): 988-995, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29550474

RESUMO

Inflammatory bowel disease (IBD) is an idiopathic disease caused by a dysregulated immune response to intestinal microbes in an individual with a genetic predisposition. Therefore, alleviation of inflammation is very important to treat IBD. Mesenchymal stem cells (MSCs) have been highlighted as new candidates for treating autoimmune disease based on their immunomodulatory properties. In this study, we investigated the anti-inflammatory mechanism and therapeutic effects of adipose tissue-derived MSCs (ASCs) using THP-1 macrophages and dextran sodium sulfate (DSS)-induced mice with chronic colitis. LPS-treated THP-1 cells expressed mRNA of CD11b, an M1 macrophage marker, at day 2. However, THP-1 co-cultured with ASCs expressed mRNA of CD206, CD68, CCL18, legumain, and IL-10, markers of M2 macrophages. In THP-1 cells co-cultured with ASCs, precursor (pro)-IL-1ß, Cox-2, and NLRP3 increased dramatically compared to LPS-treated THP-1 cells. Secretion of IL-1ß and IL-18 was significantly inhibited by ASCs, but PGE2 production was highly increased in co-culture conditions of THP-1 and ASCs. IL-18 secretion was inhibited by PGE2 treatment, and PGE2 inhibited inflammasome complex (ASC/Cas-1/NLRP3) formation in THP-1 cells. In the DSS-induced chronic colitis model, ASCs ameliorated colitis by decreasing the total number of macrophages and the M1 macrophage population. Our results suggest that ASCs can suppress the inflammatory response by controlling the macrophage population, and ASCs may be therapeutically useful for the treatment of IBD.


Assuntos
Tecido Adiposo/citologia , Colite/prevenção & controle , Dinoprostona/farmacologia , Inflamassomos/antagonistas & inibidores , Macrófagos/imunologia , Células-Tronco Mesenquimais/fisiologia , Animais , Contagem de Células , Técnicas de Cocultura , Colite/induzido quimicamente , Sulfato de Dextrana , Dinoprostona/biossíntese , Humanos , Inflamassomos/biossíntese , Macrófagos/citologia , Camundongos , Células THP-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA