Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Int J Med Sci ; 21(7): 1307-1320, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38818471

RESUMO

Transforming growth factor-ß (TGF-ß) is strongly associated with the cell adhesion signaling pathway in cell differentiation, migration, etc. Mechanistically, TGF-ß is secreted in an inactive form and localizes to the extracellular matrix (ECM) via the latent TGF-ß binding protein (LTBP). However, it is the release of mature TGF-ß that is essential for the activation of the TGF-ß signaling pathway. This progress requires specific integrins (one of the main groups of cell adhesion molecules (CAMs)) to recognize and activate the dormant TGF-ß. In addition, TGF-ß regulates cell adhesion ability through modulating CAMs expression. The aberrant activation of the TGF-ß signaling pathway, caused by abnormal expression of key regulatory molecules (such as Smad proteins, certain transcription factors, and non-coding RNAs), promotes tumor invasive and metastasis ability via epithelial-mesenchymal transition (EMT) during the late stages of tumorigenesis. In this paper, we summarize the crosstalk between TGF-ß and cell adhesion signaling pathway in cancer and its underlying molecular mechanisms.


Assuntos
Adesão Celular , Transição Epitelial-Mesenquimal , Neoplasias , Transdução de Sinais , Fator de Crescimento Transformador beta , Humanos , Fator de Crescimento Transformador beta/metabolismo , Neoplasias/patologia , Neoplasias/metabolismo , Neoplasias/genética , Transição Epitelial-Mesenquimal/genética , Integrinas/metabolismo , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica
2.
Int J Med Sci ; 21(5): 904-913, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38617002

RESUMO

Dysregulation of cellular metabolism is a key marker of cancer, and it is suggested that metabolism should be considered as a targeted weakness of colorectal cancer. Increased polyamine metabolism is a common metabolic change in tumors. Thus, targeting polyamine metabolism for anticancer therapy, particularly polyamine blockade therapy, has gradually become a hot topic. Quercetin-3-methyl ether is a natural compound existed in various plants with diverse biological activities like antioxidant and antiaging. Here, we reported that Quercetin-3-methyl ether inhibits colorectal cancer cell viability, and promotes apoptosis in a dose-dependent and time-dependent manner. Intriguingly, the polyamine levels, including spermidine and spermine, in colorectal cancer cells were reduced upon treatment of Quercetin-3-methyl ether. This is likely resulted from the downregulation of SMOX, a key enzyme in polyamine metabolism that catalyzes the oxidation of spermine to spermidine. These findings suggest Quercetin-3-methyl ether decreases cellular polyamine level by suppressing SMOX expression, thereby inducing colorectal cancer cell apoptosis. Our results also reveal a correlation between the anti-tumor activity of Quercetin-3-methyl ether and the polyamine metabolism modulation, which may provide new insights into a better understanding of the pharmacological activity of Quercetin-3-methyl ether and how it reprograms cellular polyamine metabolism.


Assuntos
Produtos Biológicos , Neoplasias Colorretais , Quercetina/análogos & derivados , Humanos , Poliaminas , Espermidina , Espermina , Apoptose , Neoplasias Colorretais/tratamento farmacológico
3.
J Transl Med ; 19(1): 356, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34407839

RESUMO

Inflammatory bowel diseases (IBD), mainly comprising ulcerative colitis (UC) and Crohn's Disease, are most often a polygenic disorder with contributions from the intestinal microbiome, defects in barrier function, and dysregulated host responses to microbial stimulation. Strategies that target the microbiota have emerged as potential therapies and, of these, probiotics have gained the greatest attention. Herein, we isolated a strain of Lactobacillus paracasei R3 (L.p R3) with strong biofilm formation ability from infant feces. Interestingly, we also found L.p R3 strain can ameliorate the general symptoms of murine colitis, alleviate inflammatory cell infiltration and inhibit Th17 while promote Treg function in murine dextran sulfate sodium (DSS)-induced colitis. Overall, this study suggested that L.p R3 strain significantly improves the symptoms and the pathological damage of mice with colitis and influences the immune function by regulating Th17/Treg cell balance in DSS-induced colitis in mice.


Assuntos
Colite , Lacticaseibacillus paracasei , Animais , Colite/induzido quimicamente , Colo , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores , Células Th17
4.
Exp Cell Res ; 395(1): 112170, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32682783

RESUMO

Colorectal cancer is the second leading cause of cancer mortality worldwide with poor prognosis and high recurrence. Aberrant Wnt/ß-catenin signaling promotes oncogenesis by transcriptional activation of c-Myc and its downstream signals. EDAR is characterized as an important effector of canonical Wnt signaling in developing skin appendages, but the interplay between EDAR and Wnt signaling in tumorigenesis and progression remains to be elucidated. In this study, we revealed that EDAR expression is prevalently elevated in colorectal cancer tissues compared with normal tissues. Further analysis suggests there is a strict correlation between EDAR expression and colorectal cancer progression. EDAR silencing by shRNA in colorectal cancer cells showed proliferative suppression via retarding cell cycle at G1 phase. Xenograft mice transplanted with shEDAR-transduced tumor cells significantly alleviated tumor burden in comparison with control mice. Furthermore, downregulation of EDAR was accompanied by reduction of ß-catenin, c-Myc and other G1 cell cycle regulators, while ß-catenin agonist restored the expression of these proteins and overrode the proliferative block induced by EDAR knockdown. These findings indicate that EDAR functions as a component of Wnt/ß-catenin signaling pathway, and is a potential modulator in colorectal carcinogenesis.


Assuntos
Proliferação de Células/fisiologia , Neoplasias do Colo , Neoplasias Colorretais/patologia , Recidiva Local de Neoplasia/metabolismo , Receptores da Ectodisplasina/metabolismo , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Recidiva Local de Neoplasia/genética , Receptores da Ectodisplasina/genética , Via de Sinalização Wnt/genética
6.
Mediators Inflamm ; 2016: 3214105, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27247488

RESUMO

Epstein-Barr virus-induced gene 3 (EBI3) is a member of the interleukin-12 (IL-12) family structural subunit and can form a heterodimer with IL-27p28 and IL-12p35 subunit to build IL-27 and IL-35, respectively. However, IL-27 stimulates whereas IL-35 inhibits antitumor T cell responses. To date, little is known about the role of EBI3 in tumor microenvironment. In this study, firstly we assessed EBI3, IL-27p28, IL-12p35, gp130, and p-STAT3 expression with clinicopathological parameters of colorectal cancer (CRC) tissues; then we evaluated the antitumor T cell responses and tumor growth with a EBI3 blocking peptide. We found that elevated EBI3 may be associated with IL-12p35, gp130, and p-STAT3 to promote CRC progression. EBI3 blocking peptide promoted antitumor cytotoxic T lymphocyte (CTL) response by inducing Granzyme B, IFN-γ production, and p-STAT3 expression and inhibited CRC cell proliferation and tumor growth to associate with suppressing gp130 and p-STAT3 expression. Taken together, these results suggest that EBI3 may mediate a bidirectional reciprocal-regulation STAT3 signaling pathway to assist the tumor escape immune surveillance in CRC.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Interleucinas/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Peptídeos/uso terapêutico , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/metabolismo , Adulto , Idoso , Animais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Interleucinas/antagonistas & inibidores , Masculino , Camundongos , Pessoa de Meia-Idade , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Mediators Inflamm ; 2016: 8026494, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27006530

RESUMO

Recent studies suggest that tumor-associated macrophage-produced IL-6 is an important mediator within the tumor microenvironment that promotes tumor growth. The activation of IL-6/STAT3 axis has been associated with chemoresistance and poor prognosis of a variety of cancers including colorectal carcinoma and thus serves as a potential immunotherapeutic target for cancer treatment. However, it is not fully understood whether anticytokine therapy could reverse chemosensitivity and enhance the suppressive effect of chemotherapy on tumor growth. In this study, we aimed to investigate the effect of IL-6 inhibition therapy on the antitumor effect of carboplatin. Enhanced expression of IL-6 and activation of STAT3 were observed in human colorectal carcinoma samples compared to normal colorectal tissue, with higher levels of IL-6/STAT3 in low grade carcinomas. Treatment of carboplatin (CBP) dose-dependently increased IL-6 production and STAT3 activation in human colorectal LoVo cells. Blockade of IL-6 with neutralizing antibody enhanced chemosensitivity of LoVo cells to carboplatin as evidenced by increased cell apoptosis. IL-6 blockade abolished carboplatin-induced STAT3 activation. IL-6 blockade and carboplatin synergistically reduced cyclin D1 expression and enhanced caspase-3 activity in LoVo cells. Our results suggest that inhibition of IL-6 may enhance chemosensitivity of colon cancers with overactive STAT3 to platinum agents.


Assuntos
Carboplatina/farmacologia , Neoplasias Colorretais/metabolismo , Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Transdução de Sinais/efeitos dos fármacos
8.
Arch Toxicol ; 87(7): 1273-85, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23423712

RESUMO

Trimethyltin chloride (TMT) has been known as a classic neurotoxicant which can cause serious neuronal degeneration diseases. Nuclear factor κB (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways play pivotal role in the central nerves system. In the present study, the intracellular pathways involved in TMT-induced apoptosis on human neuroblastoma cells SY5Y (SH-SY5Y) were investigated. We observed high level of nuclear NF-κB p65 submit, activated JNK, ERK, and p38 by TMT exposure. In contrast, low level of Bcl-2 and XIAP (two known NF-κB-regulated endogenous anti-apoptotic molecules) was present. To further investigate the role of these pathways and the relationship between them, specific inhibitors were used and the alteration of each pathway was evaluated. Pretreatment with MG132, an inhibitor of proteasome activity, and BAY11-7082, an inhibitor of IκBα phosphorylation, both inhibited NF-κB p65 translocation and significantly promoted apoptosis. NF-κB inhibition also induced down-expression of Bcl-2 and XIAP, exaggerated JNK phosphorylation, and ERK inhibition. SP600125 and U0126, by blocking the phosphorylation of c-Jun and MEK1/2, inhibited JNK and ERK phosphorylation, respectively, and attenuated apoptosis significantly. JNK and ERK inhibition also induced IκBα degradation and NF-κB p65 translocation, leading to expression of Bcl-2 and XIAP. The detrimental role of MG132 and BAY11-7082 appears related to the exaggerated JNK phosphorylation. The SP600125 and U0126 neuroprotection appears related to NF-κB-regulated transcriptional control of Bcl-2 and XIAP. These results suggest that the cross-talk and a balance between NF-κB and MAPKs may be involved in TMT-induced apoptosis on SH-SY5Y cells.


Assuntos
Apoptose/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Compostos de Trimetilestanho/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Inibidor de NF-kappaB alfa , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Neurônios/enzimologia , Neurônios/patologia , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fatores de Tempo , Fator de Transcrição RelA/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Artigo em Zh | MEDLINE | ID: mdl-23510832

RESUMO

OBJECTIVE: To investigate the cytotoxicity of bromoxynil on SH-SY5Y cells and its effect on the expression of nuclear factor-kappa B (NF-κB) and I kappa B alpha (IκBα) in SH-SY5Y cells. METHODS: SH-SY5Y cells were exposed to bromoxynil (10, 50, or 100 µmol/L) for 24 and 48 h, and other SH-SY5Y cells, which were used as a control, were exposed only to dimethyl sulfoxide. After 24 and 48 h of exposure, the morphological changes of these cells were observed under an inverted microscope, and the cytotoxicity of bromoxynil was measured by MTT assay. The cellular proliferation was examined by cell counting after 12, 24, 48, 72, and 96 h of exposure. After 24 h of exposure, the expression of NF-κB was evaluated by Western blot and immunocytochemistry, and the expression of IκBα was evaluated by Western blot. RESULTS: The cellular proliferation inhibition rates (CPIRs) of 50 and 100 µmol/L groups were significantly higher than that of the control group after 24 and 48 h of exposure (P < 0.05); the CPIR was significantly higher after 48 h than after 24 h in the two groups (P < 0.05). The growth curve revealed that these groups began to show differences in cell count at the 24th of exposure and that the differences were even more marked as the exposure went on (F = 17.15, P < 0.05). The control group had a significantly increased cell count at the 48th, 72nd, and 96th h of exposure (P < 0.05); the 10 and 50 µmol/L groups had a significantly increased cell count at the 72nd and 96th h of exposure (P < 0.05); the 100 µmol/L group showed no significant change in cell count during 96h of exposure. The 50 and 100 µmol/L groups hada significantly longer cell doubling time than the control group (P < 0.05). The immunocytochemistry showed that as the dose of bromoxynil increased, the brownish yellow particles in the cytoplasm and nuclei became darker, the expression of NF-κB was upregulated, and the nuclear translocation of NF-κB was increased. The Western blot showed that the 100 µmol/L group had significantly higher expression of NF-κB in the nuclei than the control group (P < 0.05) and that the 50 and 100 µmol/L groups had significantly lower expression of IκBα in total proteins than the control group (P < 0.05). CONCLUSION: Bromoxynil can inhibit the proliferation of SH-SY5Y cells under this experimental condition, which may be related to activation of NF-κB.


Assuntos
Proliferação de Células/efeitos dos fármacos , NF-kappa B/metabolismo , Nitrilas/toxicidade , Linhagem Celular Tumoral , Humanos , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa
10.
Cell Death Dis ; 14(8): 579, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37653021

RESUMO

Eukaryotic initiation factor 5A2 (eIF5A2) is overexpressed in many types of cancer, and spermidine-mediated eIF5A hypusination (eIF5Ahpu) appears to be essential to most of eIF5A's biological functions, including its important role in regulating cancer cell proliferation, epithelial-mesenchymal transition (EMT), and cancer stem cell (CSC) properties as well as immune cell functions. Here we investigated the role of eIF5Ahpu in the growth of oral squamous cell carcinoma cells (OSCCs) and OSCC-induced polarization of M2-like tumor-associated macrophages (TAMs). TCGA dataset analysis revealed an overall upregulation in the mRNA expression of eIF5A2 and several key enzymes involved in polyamine (PA) metabolism in HNSCC, which was confirmed by Western blot and IHC studies. Blocking eIF5Ahpu by GC-7 but not the upstream key enzyme activities of PA metabolism, remarkably inhibited cell proliferation and the expression of EMT- and CSC-related genes in OSCC cells. In addition, blocking eIF5Ahpu robustly inhibited OSCC-induced M2-like TAM polarization in vitro. More Importantly, blocking eIF5Ahpu dramatically retarded tumor growth and infiltration/polarization of M2-like TAM in a syngeneic orthotopic murine tongue SCC model. Thus, eIF5Ahpu plays dual functions in regulating tumor cell growth and polarization of M2-TAMs in OSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Bucais , Neoplasias da Língua , Animais , Camundongos , Neoplasias Bucais/genética , Fatores de Iniciação de Peptídeos/genética , Neoplasias da Língua/genética , Macrófagos Associados a Tumor , Humanos , Fator de Iniciação de Tradução Eucariótico 5A
11.
Biochemistry ; 51(16): 3523-30, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22482799

RESUMO

Transthyretin (TTR) is a cerebrospinal fluid and serum protein that undergoes ordered aggregation (amyloidogenesis) in familial amyloidotic polyneuropathy (FAP) and senile systemic amyloidosis (SSA). It is now widely accepted that dissociation of the native TTR tetramer is a precondition for amyloidogenesis; thus, molecules that stabilize the tetramer have received much attention as potential TTR amyloidosis inhibitors. Many of these inhibitors bind to the thyroxine (T(4)) binding pocket and interact specifically with a section of the TTR sequence, corresponding to residues 105-115, that is implicated in amyloidogenic propensity. In this work, we study the effects of "stabilizing" inhibitors on ordered aggregation of TTR(105-115) peptide. We show that molecules known to bind full-length TTR at the T(4) site are potent, specific inhibitors of ordered aggregation, while molecules that do not interact with TTR exhibit milder, nonspecific disruption through a "hyperbundling" effect. Our results suggest that, in addition to annealing the native tetramer, "stabilizing" inhibitors may also directly disrupt amyloidogenic aggregation of TTR monomers through specific interactions with the exposed TTR(105-115) sequence.


Assuntos
Amiloide/química , Amiloidose/metabolismo , Pré-Albumina/química , Amiloide/metabolismo , Neuropatias Amiloides Familiares/metabolismo , Humanos , Cinética , Peptídeos/química , Peptídeos/metabolismo , Pré-Albumina/antagonistas & inibidores , Pré-Albumina/metabolismo , Dobramento de Proteína
12.
Inorg Chem ; 51(1): 674-9, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22141338

RESUMO

A Mn(II)-based homometallic porous metal-organic framework, Mn(5)(btac)(4)(µ(3)-OH)(2)(EtOH)(2)·DMF·3EtOH·3H(2)O (1, btac = benzotriazole-5-carboxylate), has been solvothermally synthesized and structurally characterized by elemental analysis, thermogravimetric analysis, and X-ray crystallographic study. 1 is a 3D neutral framework featuring 1D porous channels constructed by {Mn-OH-Mn}(n) chains and btac linkers. Magnetic studies show that 1 is a 3D metamagnet containing 1D {Mn-OH-Mn}(n) ferrimagnetic chains. High-pressure H(2) adsorption measurement at 77 K reveals that activated 1 can absorb 0.99 wt % H(2) at 0.5 atm and reaches a maximum of 1.03 wt % at 5.5 atm. The steep H(2) absorption at lower pressure (98.2% of the storage capacity at 0.5 atm) is higher than the corresponding values of some MOFs (MIL-100 (16.1%), MOF-177 (57.1%), and MOF-5 (22.2%)). Furthermore, activated 1 can adsorb CO(2) at room temperature and 275 K. The adsorption enthalpy is 22.0 kJ mol(-1), which reveals the high binding ability for CO(2). Detailed gas sorption implies that the exposed Mn(II) coordination sites in the activated 1 play an important role to improve its adsorption capacities.


Assuntos
Hidrogênio/isolamento & purificação , Imãs/química , Manganês/química , Compostos Organometálicos/química , Triazóis/química , Adsorção , Ácidos Carboxílicos/síntese química , Ácidos Carboxílicos/química , Cristalografia por Raios X , Modelos Moleculares , Compostos Organometálicos/síntese química , Porosidade , Pressão , Triazóis/síntese química
13.
Artigo em Zh | MEDLINE | ID: mdl-23257029

RESUMO

OBJECTIVE: To investigate the effects of trimethyltin chloride (TMT) on proliferation, apoptosis, oxidative damage, and NF-κB expression in PC12 cells in vitro. METHODS: PC12 cells were treated with 0, 0.3125, 0.6250, 1.2500, 2.5000, 5.0000, 10.0000, and 20.0000 µmol/L TMT for 24 and 48 h, and MTT assay was used to evaluate cell viability. PC12 cells were treated with 1.25, 2.50, 5.00, and 10.00 µmol/L TMT for 12 and 24 h, and flow cytometry was used to measure the apoptotic rates of cells. PC12 cells were treated with 1.25, 2.50, 5.00, and 10.00 µmol/L TMT for 6 h, and the reactive oxygen species (ROS) and glutathione (GSH) levels were measured. PC12 cells were treated with 1.25, 2.50, 5.00, and 10.00 µmol/L TMT for 12 h, and Western blot was used to measure NF-κB levels. RESULTS: Compared with solvent controls, the PC12 cells treated with 2.5000, 5.0000, 10.0000, and 20.0000 µmol/L TMT for 24 h showed significantly decreased cell viability (P < 0.05); the PC12 cells treated with 1.2500, 2.5000, 5.0000, 10.0000, and 20.0000 µmol/L TMT for 48 h showed significantly decreased cell viability (P < 0.05). The PC12 cells treated with 1.2500, 2.5000, 5.0000, and 10.0000 µmol/L TMT for 12 h had apoptotic rates of 15.30% ± 0.75%, 18.90% ± 0.61%, 22.00% ± 0.60%, and 36.50% ± 0.66%, respectively, and the PC12 cells treated with 1.25, 2.50, 5.00, and 10.00 µmol/L TMT for 24 h had apoptotic rates of 28.6% ± 0.40%, 43.54% ± 2.00%, 65.73% ± 0.71%, and 74.67% ± 0.40%, respectively, all significantly higher than those of the control group (12 h: 12.80% ± 1.00%, 24h: 16.83% ± 0.25%) (P < 0.05). The ROS fluorescence intensities of the PC12 cells treated with 1.25, 2.50, 5.00, and 10.00 µmol/L TMT were 1.42, 1.71, 1.78, and 1.89 times that of the control group (P < 0.05); the PC12 cells treated with 2.50, 5.00, and 10.00 µmol/L TMT had GSH levels of 0.17 ± 0.0, 0.20 ± 0.04, and 0.07 ± 0.03 µmol/µg protein, significantly lower than that of the control group (0.30 ± 0.01 µmol/L protein) (P < 0.05). The PC12 cells treated with 2.50, 5.00, and 10.00 µmol/L TMT had significantly higher expression of NF-κB p65 than the control group (P < 0.05). CONCLUSION: Under our laboratory conditions, TMT can significantly inhibit proliferation and induce apoptosis in PC12 cells, which may be related to oxidative stress and NF-κB signaling pathway activation.


Assuntos
Apoptose/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Compostos de Trimetilestanho/toxicidade , Animais , Proliferação de Células/efeitos dos fármacos , Células PC12 , Ratos , Fator de Transcrição RelA/metabolismo
14.
Front Oncol ; 12: 860932, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35719975

RESUMO

Breast cancer has become the most common malignant disease in the world according to the International Agency for Research on Cancer (IARC), and the most critical cause of death is distant metastasis. The lung is the extremely common visceral site for breast cancer metastasis. Lung metastasis of breast cancer is not only dependent on the invasive ability of the tumor itself, but also closely relates to the pulmonary microenvironment. In the progression of breast cancer, the formation of specific microenvironment in lungs can provide suitable conditions for the metastasis of breast cancer. Pulmonary inflammatory response, angiogenesis, extracellular matrix remodeling, some chemotherapeutic agents and so on all play important roles in the formation of the pulmonary microenvironment. This review highlights recent findings regarding the alterations of pulmonary microenvironment in lung metastasis of breast cancer, with a focus on various cells and acellular components.

15.
Front Immunol ; 13: 952071, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35990688

RESUMO

Inflammatory bowel diseases (IBDs) represent a group of chronic inflammatory disorders of the gastrointestinal (GI) tract including ulcerative colitis (UC), Crohn's disease (CD), and unclassified IBDs. The pathogenesis of IBDs is related to genetic susceptibility, environmental factors, and dysbiosis that can lead to the dysfunction of immune responses and dysregulated homeostasis of local mucosal tissues characterized by severe inflammatory responses and tissue damage in GI tract. To date, extensive studies have indicated that IBDs cannot be completely cured and easy to relapse, thus prompting researchers to find novel and more effective therapeutics for this disease. Due to their potent multipotent differentiation and immunomodulatory capabilities, mesenchymal stem/stromal cells (MSCs) not only play an important role in regulating immune and tissue homeostasis but also display potent therapeutic effects on various inflammatory diseases, including IBDs, in both preclinical and clinical studies. In this review, we present a comprehensive overview on the pathological mechanisms, the currently available therapeutics, particularly, the potential application of MSCs-based regenerative therapy for IBDs.


Assuntos
Colite Ulcerativa , Doença de Crohn , Doenças Inflamatórias Intestinais , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Doença de Crohn/terapia , Humanos , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/terapia
16.
Front Immunol ; 13: 912279, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36119047

RESUMO

The study of metabolism provides important information for understanding the biological basis of cancer cells and the defects of cancer treatment. Disorders of polyamine metabolism is a common metabolic change in cancer. With the deepening of understanding of polyamine metabolism, including molecular functions and changes in cancer, polyamine metabolism as a new anti-cancer strategy has become the focus of attention. There are many kinds of polyamine biosynthesis inhibitors and transport inhibitors, but not many drugs have been put into clinical application. Recent evidence shows that polyamine metabolism plays essential roles in remodeling the tumor immune microenvironment (TIME), particularly treatment of DFMO, an inhibitor of ODC, alters the immune cell population in the tumor microenvironment. Tumor immunosuppression is a major problem in cancer treatment. More and more studies have shown that the immunosuppressive effect of polyamines can help cancer cells to evade immune surveillance and promote tumor development and progression. Therefore, targeting polyamine metabolic pathways is expected to become a new avenue for immunotherapy for cancer.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/farmacologia , Humanos , Imunidade , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Poliaminas/metabolismo , Microambiente Tumoral
17.
Stem Cells Int ; 2022: 6544514, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35813890

RESUMO

Human gingiva-derived mesenchymal stem cells (GMSCs) are isolated from the gingival propria with promising regenerative, immunomodulatory, and anti-inflammatory properties. Recently, several studies, including ours, have found that GMSCs have the therapeutic potentials of nerve regeneration and skin disorders in various types such as the cell itself, cell-free conditioned medium, or extracellular vesicles (EVs). However, the mechanobiological behavior of GMSCs is closely related to the culture conditions. Therefore, the purpose of this study was to evaluate the function of human GMSCs on imiquimod- (IMQ-) induced murine psoriasis-like skin inflammation in two-dimensional (2D) and three-dimensional (3D) culture conditions. Here, we isolated and characterized GMSCs in 2D and 3D culture conditions and found that GMSCs in 2D and 3D infusion can significantly ameliorate the IMQ-induced murine psoriasis-like skin inflammation, reduce the levels of Th1- and Th17-related cytokines IFN-γ, TNF-α, IL-6, IL-17A, IL-17F, IL-21, and IL-22, and upregulate the percentage of spleen CD25+CD3+ T cells while downregulate the percentage of spleen IL-17+CD3+ T cells. In summary, our novel findings reveal that GMSCs in 2D and 3D infusion may possess therapeutic effects in the treatment of psoriasis.

18.
Front Surg ; 9: 1089244, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36969323

RESUMO

Low back pain is one of the top disorders that leads to disability and affects disability-adjusted life years (DALY) globally. Intervertebral disc degeneration (IDD) and subsequent discogenic pain composed major causes of low back pain. Recent studies have identified several important risk factors contributing to IDD's development, such as inflammation, mechanical imbalance, and aging. Based on these etiology findings, three categories of animal models for inducing IDD are developed: the damage-induced model, the mechanical model, and the spontaneous model. These models are essential measures in studying the natural history of IDD and finding the possible therapeutic target against IDD. In this review, we will discuss the technical details of these models, the duration between model establishment, the occurrence of observable degeneration, and the potential in different study ranges. In promoting future research for IDD, each animal model should examine its concordance with natural IDD pathogenesis in humans. We hope this review can enhance the understanding and proper use of multiple animal models, which may attract more attention to this disease and contribute to translation research.

19.
J Cancer ; 12(1): 150-162, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33391411

RESUMO

Anaplastic lymphoma kinase (ALK) has been described in a range of human cancers and is involved in cancer initiation and progression via activating multiple signaling pathways, such as the PI3K-AKT, CRKL-C3G, MEKK2/3-MEK5-ERK5, JAK-STAT and MAPK signal pathways. Recently ALK and LTK ligand 1 (ALKAL1) also named "augmentor-ß" or "FAM150A" is identified as a potent activating ligands for human ALK that bind to the extracellular domain of ALK. However, due to its poor stability, the mechanisms of ALKAL1 underlying the tumor progression in the human cancers including colorectal cancer have not been well documented. Herein, ALKAL1 expression was evaluated by RNA sequencing datasets from The Cancer Genome Atlas (TCGA) of 625 cases colorectal cancer, immunohistochemical analysis of 377 cases colorectal cancer tissues, and Western blotting even Real-time PCR of 10 pairs of colorectal cancer tissues and adjacent normal tissues, as well as 8 colorectal cancer cell lines. Statistical analysis was performed to explore the correlation between ALKAL1 expression and clinicopathological features in colorectal cancer. Univariate and multivariate Cox regression analysis were performed to examine the association between ALKAL1 expression and overall survival. In vitro and in vivo assays were performed to assess the biological roles of ALKAL1 in colorectal cancer. Gene set enrichment analysis (GSEA), Western blotting and luciferase assays were used to identify the underlying signal pathway involved in the tumor progression role of ALKAL1. As a result, we showed that ALKAL1 was upregulated in colorectal cancer tissues and cell lines. Upregulation of ALKAL1 correlated with tumor malignancy and poor prognosis in colorectal cancer. ALKAL1 silencing inhibited tumorigenesis, metastasis and invasion of colorectal cancer cells, and inhibited SHH signaling pathway, which is essential for ALKAL1 induced migration. Our findings reveal a new mechanism by which ALKAL1 participates in colorectal cancer migration and invasion via activating the SHH signaling pathway.

20.
Ann Transl Med ; 9(12): 1016, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34277816

RESUMO

BACKGROUND: Beta-1 syntrophin (SNTB1) is an intracellular scaffold protein that provides a platform for the formation of signal transduction complexes, thereby modulating and coordinating various intracellular signaling events and crucial cellular processes. However, the physiological role of SNTB1 is poorly understood. This study aims to explore the role of SNTB1 in colorectal cancer (CRC) tumorigenesis and progression, with particular focus on SNTB1's expression pattern, clinical relevance, and possible molecular mechanism in CRC development. METHODS: SNTB1 expression was analyzed in both clinical tissues and The Cancer Genome Atlas (TCGA) database. Real-time polymerase chain reaction (PCR), Western blot, and immunohistochemical assays were used to detect the relative mRNA and protein levels of SNTB1. Statistical analysis was performed to examine the correlation between SNTB1 expression and the clinicopathological characteristics of patients with CRC. Bioinformatics gene set enrichment analysis (GSEA), Western blot, luciferase assay, and agonist recovery assays were conducted to evaluate the relevance of SNTB1 and the ß-catenin signaling pathway in CRC. A flow cytometry-based Hoechst 33342 efflux assay was applied to assess the proportion of the side population (SP) within total CRC cells. RESULTS: Elevated levels of SNTB1 were identified in CRC tissues and cell lines. The elevation of SNTB1 was positively correlated with the degree of malignancy and poor prognosis in CRC. We further revealed that, by modulating the ß-catenin signaling pathway, silencing SNTB1 expression suppressed tumor growth and cancer stemness in vitro, as well as tumorigenesis in vivo. CONCLUSIONS: These findings suggest that SNTB1 plays a crucial role in colorectal tumorigenesis and progression by modulating ß-catenin signaling and the stemness maintenance of cancer cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA