Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int Wound J ; 21(4): e14807, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38591163

RESUMO

Skin Cutaneous Melanoma (SKCM) is a form of cancer that originates in the pigment-producing cells, known as melanocytes, of the skin. Delay wound healing is often correlated with the occurrence of and progression of SKCM. In this comprehensive study, we investigated the intricate roles of two important wound healing genes in SKCM, including Matrix Metalloproteinase-2 (MMP2) and Matrix Metalloproteinase-9 (MMP9). Through a multi-faceted approach, we collected clinical samples, conducted molecular experiments, including RT-qPCR, bisulphite sequencing, cell culture, cell Counting Kit-8, colony formation, and wound healing assays. Beside this, we also used various other databases/tools/approaches for additional analysis including, UALCAN, GEPIA, HPA, MEXPRESS, cBioPortal, KM plotter, DrugBank, and molecular docking. Our results revealed a significant up-regulation of MMP2 and MMP9 in SKCM tissues compared to normal counterparts. Moreover, promoter methylation analysis suggested an epigenetic regulatory mechanism. Validations using TCGA datasets and immunohistochemistry emphasized the clinical relevance of MMP2 and MMP9 dysregulation. Functional assays demonstrated their synergistic impact on proliferation and migration in SKCM cells. Furthermore, we identified potential therapeutic candidates, Estradiol and Calcitriol, through drug prediction and molecular docking analyses. These compounds exhibited binding affinities, suggesting their potential as MMP2/MMP9 inhibitors. Overall, our study elucidates the diagnostic, prognostic, and therapeutic implications of MMP2 and MMP9 in SKCM, shedding light on their complex interplay in SKCM occurrence and progression.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/genética , Melanoma/terapia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/terapia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz , Simulação de Acoplamento Molecular , Cicatrização/genética , Mutação , Metilação
2.
J Neurotrauma ; 40(13-14): 1402-1414, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36994490

RESUMO

Abstract Making an appropriate diagnosis and administering effective treatment for hydrocephalus in patients with severe disorders of consciousness (DOC) remains controversial and difficult. Given that the typical symptoms are usually concealed by the limited behavioral responsiveness of patients with severe DOC, hydrocephalus diagnosis is likely to be missed in the clinic. Even if not, the presence of hydrocephalus may reduce the likelihood of DOC recovery, posing a conundrum for clinicians. From December 2013 to January 2023, the clinical data and therapeutic schedule of hydrocephalus in patients with severe DOC at Huashan Hospital's Neurosurgical Emergency Center were studied retrospectively. Sixty-eight patients (mean age [± SD] 52.53 ± 17.03 years, 35 males and 33 females) with severe DOC were included. The hydrocephalus was discovered after computed tomography (CT) or magnetic resonance imaging (MRI) revealed enlarged ventricles in the patients. During hospitalization, patients underwent a surgical treatment that included a ventriculoperitoneal (V-P) shunt and/or cranioplasty (CP) implantation. Following the surgery, an individualized V-P pressure was established based on the patient's ventricle size and neurological function variation. To account for the improvement in consciousness in patients with severe DOC, Glasgow Coma Scale (GCS) and Coma Recovery Scale-Revised (CRS-R) assessments were performed before and after hydrocephalus treatment. All patients with severe DOC had varying degrees of ventricular enlargement, deformation, and poor brain compliance. Approximately 60.3% (41/68) of them had low- or negative-pressure hydrocephalus (LPH or NegPH). Of the patients, 45.5% (31/68) had a one-stage V-P shunt and CP operation performed concurrently, whereas the remaining 37 patients had a single V-P shunt operation performed independently. Besides two patients with DOC who developed surgical complications, 92.4% (61/66) of the survivors showed an improvement in consciousness after hydrocephalus treatment. In patients with severe DOC, LPH or NegPH was common. Secondary hydrocephalus in patients with DOC had been largely ignored, hampering their neurological rehabilitation. Even months or years after the onset of severe DOC, active treatment of hydrocephalus can significantly improve patients' consciousness and neurological function. This study summarized several evidence-based treatment experiences of hydrocephalus in patients with DOC.


Assuntos
Transtornos da Consciência , Hidrocefalia , Masculino , Feminino , Humanos , Adulto , Pessoa de Meia-Idade , Idoso , Estudos Retrospectivos , Transtornos da Consciência/etiologia , Transtornos da Consciência/terapia , Transtornos da Consciência/diagnóstico , Estado de Consciência , Hidrocefalia/diagnóstico por imagem , Hidrocefalia/etiologia , Hidrocefalia/cirurgia , Resultado do Tratamento
3.
Oncol Rep ; 49(2)2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36579667

RESUMO

The epidermal growth factor receptor (EGFR) remains one of the best molecules for developing targeted therapy for multiple human malignancies, including head and neck squamous cell carcinoma (HNSCC). Small molecule inhibitors or antibodies targeting EGFR have been extensively developed in recent decades. Immunotoxin (IT)­based therapy, which combines cell surface binding ligands or antibodies with a peptide toxin, represents another cancer treatment option. A total of 3 diphtheria toxin (DT)­based fusion toxins that target human EGFR­monovalent EGFR IT (mono­EGF­IT), bivalent EGFR IT (bi­EGF­IT), and a bispecific IT targeting both EGFR and interleukin­2 receptor (bis­EGF/IL2­IT) were recently generated by the authors. Improved efficacy and reduced toxicity of bi­EGF­IT compared with mono­EGF­IT in immunocompromised HNSCC mouse models was reported. In the present study, bis­EGF/IL2­IT were generated using a unique DT­resistant yeast expression system and evaluated the in vitro and in vivo efficacy and toxicity of the 3 EGF­ITs in immunocompetent mice. The results demonstrated that while the three EGF­ITs had different efficacies in vitro and in vivo against HNSCC, bi­EGF­IT and bis­EGF/IL2­IT had significantly improved in vivo efficacy and remarkably less off­target toxicity compared with mono­EGF­IT. In addition, bis­EGF/IL2­IT was superior to bi­EGF­IT in reducing tumor size and prolonging survival in the metastatic model. These data suggested that targeting either the tumor immune microenvironment or enhancing the binding affinity could improve the efficacy of IT­based therapy. Bi­EGF­IT and bis­EGF/IL2­IT represent improved candidates for IT­based therapy for future clinical development.


Assuntos
Neoplasias de Cabeça e Pescoço , Interleucina-2 , Humanos , Animais , Camundongos , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Toxina Diftérica/farmacologia , Microambiente Tumoral
4.
Curr Med Sci ; 42(6): 1248-1255, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36542322

RESUMO

OBJECTIVE: Thyroid hormones (THs) regulate multiple physiological activities in the liver, including cellular metabolism, differentiation, and cell growth, and play important roles in the pathogenesis of hepatocellular carcinoma (HCC). Thyroid peroxidase (TPO) is a key molecule involved in the THs synthesis and signaling pathway. As an epigenetic modification, DNA methylation has a critical role in tumorigenesis with diagnostic potential. However, the connection between THs and DNA methylation has been rarely investigated. METHODS: The methylation of key TH-related genes was analyzed by in-house epigenome-wide scanning, and we further analyzed the methylation levels of the TPO promotor in 164 sample pairs of HCC and adjacent non-cancerous tissues by Sequenom EpiTYPER assays, and evaluated their clinical implications. RESULTS: We identified that the methylation of the TPO promoter was downregulated in the HCC tissues (P<0.0001) with a mean difference ranging from 18.5% to 22.3%. This methylation pattern correlated with several clinical factors, including a multi-satellite tumor, fibrous capsule, and the presence of tumor thrombus. The receiver operator characteristic (ROC) curve analysis further confirmed that the percent methylated reference (PMR) values for TPO were predictive of the tumor [the area under the curve (AUC) ranged from 0.755 to 0.818] and the thrombosis in the HCC patients (the AUC ranged from 0.706 to 0.777). CONCLUSION: These findings demonstrated that epigenetic alterations of TPO, as indicated by the PMR values, were a potential biomarker for HCC patients with tumor thrombosis.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Metilação de DNA/genética , Neoplasias Hepáticas/metabolismo , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo
5.
Mol Oncol ; 15(4): 1054-1068, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33540470

RESUMO

Epidermal growth factor receptor (EGFR) is often overexpressed in head and neck squamous cell carcinoma (HNSCC) and represents a top candidate for targeted HNSCC therapy. However, the clinical effectiveness of current Food and Drug Administration (FDA)-approved drugs targeting EGFR is moderate, and the overall survival rate for HNSCC patients remains low. Therefore, more effective treatments are urgently needed. In this study, we generated a novel diphtheria toxin-based bivalent human epidermal growth factor fusion toxin (bi-EGF-IT) to treat EGFR-expressing HNSCC. Bi-EGF-IT was tested for in vitro binding affinity, cytotoxicity, and specificity using 14 human EGFR-expressing HNSCC cell lines and three human EGFR-negative cancer cell lines. Bi-EGF-IT had increased binding affinity for EGFR-expressing HNSCC compared with the monovalent version (mono-EGF-IT), and both versions specifically depleted EGFR-positive HNSCC, but not EGFR-negative cell lines, in vitro. Bi-EGF-IT exhibited a comparable potency to that of the FDA-approved EGFR inhibitor, erlotinib, for inhibiting HNSCC tumor growth in vivo using both subcutaneous and orthotopic HNSCC xenograft mouse models. When tested in an experimental metastasis model, survival was significantly longer in the bi-EGF-IT treatment group than the erlotinib treatment group, with a significantly reduced number of metastases compared with mono-EGF-IT. In addition, in vivo off-target toxicities were significantly reduced in the bi-EGF-IT treatment group compared with the mono-EGF-IT group. These results demonstrate that bi-EGF-IT is more effective and markedly less toxic at inhibiting primary HNSCC tumor growth and metastasis than mono-EGF-IT and erlotinib. Thus, the novel bi-EGF-IT is a promising drug candidate for further development.


Assuntos
Toxina Diftérica/uso terapêutico , Fator de Crescimento Epidérmico/uso terapêutico , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Cloridrato de Erlotinib/uso terapêutico , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Oncol ; 14(5): 991-1000, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32107846

RESUMO

The majority of clinically diagnosed cutaneous T-cell lymphomas (CTCL) highly express the cell-surface markers CC chemokine receptor 4 (CCR4) and/or CD25. Recently, we have developed diphtheria toxin-based recombinant Ontak®-like human IL2 fusion toxin (IL2 fusion toxin) and anti-human CCR4 immunotoxin (CCR4 IT). In this study, we first compared the efficacy of the CCR4 IT vs IL2 fusion toxin for targeting human CD25+ CCR4+ CTCL. We demonstrated that CCR4 IT was more effective than IL2 fusion toxin. We further constructed an IL2-CCR4 bispecific IT. The bispecific IT was significantly more effective than either IL2 fusion toxin or CCR4 IT alone. The bispecific IT is a promising novel targeted therapeutic drug candidate for the treatment of refractory and recurrent human CD25+ and/or CCR4+ CTCL.


Assuntos
Toxina Diftérica/farmacologia , Imunotoxinas/farmacologia , Interleucina-2/farmacologia , Linfoma Cutâneo de Células T/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Toxina Diftérica/genética , Citometria de Fluxo , Humanos , Imunotoxinas/genética , Concentração Inibidora 50 , Interleucina-2/genética , Interleucina-2/uso terapêutico , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Linfoma Cutâneo de Células T/mortalidade , Camundongos , Receptores CCR4/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nat Prod Res ; 34(17): 2474-2481, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30602312

RESUMO

A new ocotillol-type ginsenoside, namely 12-one-pseudoginsenoside F11 (12-one-PF11), was isolated from stems and leaves of Panax quinquefolium, whose structure was elucidated 6-O-[α-L-rhamnopyranosyl-(1-2)-ß-D-glucopyranosyl]-dammar-12-one-20S,24R-epoxy-3ß,6α,25-triol. 12-one-PF11 significantly suppressed hydrogen peroxide induced oxidative stress in human lung carcinoma A549 cells. As compared with model group, 12-one-PF11 improved cell viability of A549 cells in a dose-dependent manner, and significantly decreased the generation of malondialdehyde (MDA) and increased production of superoxide dismutase (SOD) and glutathione (GSH) and protein expression levels of nuclear related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in A549 cells.


Assuntos
Antioxidantes/isolamento & purificação , Ginsenosídeos/isolamento & purificação , Estresse Oxidativo/efeitos dos fármacos , Panax/química , Células A549/efeitos dos fármacos , Células A549/metabolismo , Antioxidantes/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Ginsenosídeos/metabolismo , Humanos , Peróxido de Hidrogênio , Estrutura Molecular , Folhas de Planta/química , Caules de Planta/química
8.
BMC Neurosci ; 20(1): 63, 2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31870319

RESUMO

BACKGROUND: Autophagy is considered to be another restorative focus for the treatment of brain tumors. Although several research have demonstrated that melatonin induces autophagy in colon cancer and hepatoma cells, there has not been any direct evidence of whether melatonin is capable of inducing autophagy in human glioma cells. RESULTS: In the present research, we report that melatonin or its agonist, agomelatine, induced autophagy in A172 and U87-MG glioblastoma cells for a concentration-and time-dependent way, which was significantly attenuated by treatment with luzindole, a melatonin receptor antagonist. Furthermore, by suppressing autophagy at the late-stage with bafilomycin A1 and early stage with 3-MA, we found that the melatonin-induced autophagy was activated early, and the autophagic flux was complete. Melatonin treatment alone did not induce any apoptotic changes in the glioblastoma cells, as measured by flow cytometry. Western blot studies confirmed that melatonin alone prominently upregulated the levels of Beclin 1 and LC3 II, which was accompanied by an increase in the expression of Bcl-2, whereas it had no effect on the expression of Bax in the glioblastoma cells. Remarkably, co-treatment with 3-MA and melatonin significantly enhanced the apoptotic cell population in the glioblastoma cells, along with a prominent decrease in the expression of bcl-2 and increase in the Bax expression levels, which collectively indicated that the disruption of autophagy triggers the melatonin-induced apoptosis in glioblastoma cells. CONCLUSIONS: These results provide information indicating that melatonin may act as a common upstream signal between autophagy and apoptosis, which may lead to the development of new therapeutic strategies for glioma.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Melatonina/farmacologia , Acetamidas/farmacologia , Apoptose/fisiologia , Autofagia/fisiologia , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Glioblastoma/fisiopatologia , Humanos , Macrolídeos/farmacologia , Melatonina/agonistas , Receptores de Melatonina/agonistas , Receptores de Melatonina/antagonistas & inibidores , Triptaminas/farmacologia
9.
Phytomedicine ; 61: 152862, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31048124

RESUMO

BACKGROUND: Ginsenoside Rh2 (Rh2), an important ingredient from Panax ginseng, has received much attention due to a range of pharmacological actions. PURPOSE: The aim of the study was to investigate the therapeutic potential Rh2 on cisplatin (CDDP)-induced nephrotoxicity and to elucidate involved mechanisms. STUDY DESIGN: An in vivo mice model of CDDP-induced nephrotoxicity was established by a single intraperitoneal injection of CDDP (20 mg/kg) to assess the effects of Rh2 on renal biochemical parameter, oxidative stress, inflammation tubular cell apoptosis and serum metabolic profiles. RESULTS: Rh2 protected against CDDP-induced renal dysfunction and ameliorated CDDP-induced oxidative stress, histopathological damage, inflammation and tubular cell apoptosis in kidney. Rh2 treatment had significantly increased expression of Bcl-2 and decreased expression of p53, Bax, cytochrome c, caspase-8, caspase-9, and caspase-3 in kidney tissues. Metabolomic analysis identified 29 altered serum metabolites in Rh2 treatment mice. CONCLUSION: These results suggest that Rh2 protects against CDDP-induced nephrotoxicity via action on caspase-mediated pathway.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Apoptose/efeitos dos fármacos , Cisplatino/efeitos adversos , Ginsenosídeos/farmacologia , Rim/efeitos dos fármacos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Animais , Antineoplásicos/efeitos adversos , Caspases/metabolismo , Suplementos Nutricionais , Ginsenosídeos/química , Rim/patologia , Masculino , Espectrometria de Massas/métodos , Metaboloma/efeitos dos fármacos , Camundongos Endogâmicos ICR , Nefrite/induzido quimicamente , Nefrite/tratamento farmacológico , Nefrite/patologia , Estresse Oxidativo/efeitos dos fármacos , Panax/química
10.
Molecules ; 24(6)2019 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-30909565

RESUMO

Panax ginseng Meyer cv. Silvatica (PGS), which is also known as "Lin-Xia-Shan-Shen" or "Zi-Hai" in China, is grown in forests and mountains by broadcasting the seeds of ginseng and is harvested at the cultivation age of 15⁻20 years. In this study, four new dammarane-type triterpenoids, ginsengenin-S1 (1), ginsengenin-S2 (2), ginsenoside-S3 (3), ginsenoside-S4 (4), along with one known compound were isolated from pearl knots of PGS. Ginsengenin-S2 significantly alleviated oxidative damage when A549 cells were exposed to cigarette smoke (CS) extract. In addition, ginsengenin-S2 could inhibit the CS-induced inflammatory reaction in A549 cells. Protective effects of ginsengenin-S2 against CS-mediated oxidative stress and the inflammatory response in A549 cells may involve the Nrf2 and HDAC2 pathways.


Assuntos
Ginsenosídeos/farmacologia , Panax/química , Triterpenos/farmacologia , Células A549/efeitos dos fármacos , China , Fumar Cigarros/efeitos adversos , Ginsenosídeos/química , Humanos , Inflamação/tratamento farmacológico , Inflamação/patologia , Estresse Oxidativo/efeitos dos fármacos , Sementes/química , Triterpenos/química , Damaranos
11.
Molecules ; 24(5)2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30823679

RESUMO

Suppressive effects of ginsenoside Rh2 (Rh2), (24R)-pseudo-ginsenoside HQ (R-PHQ), and (24S)-pseudo-ginsenoside HQ (S-PHQ) against lipopolysaccharide (LPS)-induced depression-like behavior were evaluated using the forced swimming test (FST) and tail suspension test (TST) in mice. Pretreatment with Rh2, R-PHQ, and S-PHQ significantly decreased immobility time in FST and TST with clear dose-dependence, and significantly downregulated levels of serum tumor necrosis factor-α and interleukin-6, and upregulated superoxide dismutase activity in the hippocampus of LPS-challenged mice. Furthermore, R-PHQ and S-PHQ significantly increased the expression of the brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase B (TrkB), sirtuin type 1 (Sirt1), and nuclear-related factor 2, and inhibited the phosphorylation of inhibitor of κB-α and nuclear factor-κB (NF-κB) in the hippocampus of LPS-challenged mice. Additionally, the antidepressant-like effect of R-PHQ was found related to the dopaminergic (DA), γ-aminobutyric acid (GABA)ergic, and noradrenaline systems, while the antidepressive effect of S-PHQ was involved in the DA and GABAergic systems. Taken together, these results suggested that Rh2, R-PHQ, and S-PHQ produced significant antidepressant-like effects, which may be related to the BDNF/TrkB and Sirt1/NF-κB signaling pathways.


Assuntos
Antidepressivos/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Ginsenosídeos/administração & dosagem , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Depressão/induzido quimicamente , Depressão/genética , Depressão/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Elevação dos Membros Posteriores , Humanos , Interleucina-6/genética , Lipopolissacarídeos/toxicidade , Camundongos , NF-kappa B/genética , Transdução de Sinais/efeitos dos fármacos , Natação , Triterpenos/administração & dosagem , Fator de Necrose Tumoral alfa/genética
12.
Neuroimage Clin ; 22: 101741, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30878611

RESUMO

Resting-state functional MRI (R-fMRI) research has recently entered the era of "big data", however, few studies have provided a rigorous validation of the physiological underpinnings of R-fMRI indices. Although studies have reported that various neuropsychiatric disorders exhibit abnormalities in R-fMRI measures, these "biomarkers" have not been validated in differentiating structural lesions (brain tumors) as a concept proof. We enrolled 60 patients with intracranial tumors located in the unilateral cranialcavity and 60 matched normal controls to test whether R-fMRI indices can differentiate tumors, which represents a prerequisite for adapting such indices as biomarkers for neuropsychiatric disorders. Common R-fMRI indices of tumors and their counterpart control regions, which were defined as the contralateral normal areas (for amplitude of low frequency fluctuations (ALFF), fractional ALFF (fALFF), regional homogeneity (ReHo) and degree centrality (DC)) and ipsilateral regions surrounding the tumors (for voxel-mirrored homotopic connectivity (VMHC)), were comprehensively assessed. According to robust paired t-tests with a Bonferroni correction, only VMHC (Fisher's r-to-z transformed) could successfully differentiate substantial tumors from their counterpart normal regions in patients. Furthermore, ALFF and DC were not able to differentiate tumor from normal unless Z-standardization was employed. To validate the lower power of the between-subject design compared to the within-subject design, each metric was calculated in a matched control group, and robust two-sample t-tests were used to compare the patient tumors and the normal controls at the same place. Similarly, only VMHC succeeded in differentiating significant differences between tumors and the sham tumor areas of normal controls. This study tested the premise of R-fMRI biomarkers for differentiating lesions, and brings a new understanding to physical significance of the Z-standardization.


Assuntos
Mapeamento Encefálico/métodos , Neoplasias Encefálicas/diagnóstico por imagem , Cerebelo/diagnóstico por imagem , Cerebelo/fisiologia , Imageamento por Ressonância Magnética/métodos , Adulto , Idoso , Neoplasias Encefálicas/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
13.
Int J Mol Sci ; 20(4)2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30769948

RESUMO

(24R)-pseudo-ginsenoside HQ (R-PHQ) and (24S)-pseudo-ginsenoside HQ (S-PHQ) are the main metabolites of (20S)-ginsenoside Rh2 (Rh2) in vivo. In this study, we found that Rh2, R-PHQ, and S-PHQ upregulated the innate and adaptive immune response in cyclophosphamide (CTX) induced-immunocompromised mice as evidenced by the number of leukocytes, cellular immunity, and phagocytosis of macrophages. Spleen T-lymphocyte subpopulations and the serum cytokines level were also balanced in these immunosuppressed mice. Furthermore, co-administration with R-PHQ or S-PHQ did not compromise the antitumor activity of CTX in the hepatoma H22-bearing mice. Treatment with R-PHQ and S-PHQ clearly induced the apoptosis of tumor cells, significantly increased the expression of Bax, and remarkably inhibited the expression of Bcl-2 and vascular endothelial growth factor (VEGF) in H22 tumor tissues. The anti-tumor activity of R-PHQ and S-PHQ could be related to the promotion of tumor apoptosis and inhibition of angiogenesis and may involve the caspase and VEGF signaling pathways. This study provides a theoretical basis for further study on R-PHQ and S-PHQ.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Ginsenosídeos/administração & dosagem , Imunidade Inata/imunologia , Neovascularização Patológica/imunologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/imunologia , Proliferação de Células/efeitos dos fármacos , Ciclofosfamida/administração & dosagem , Humanos , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/química , Camundongos , Neovascularização Patológica/tratamento farmacológico , Fagocitose , Transdução de Sinais/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
14.
Molecules ; 23(11)2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30469321

RESUMO

In this study, the protective effects of pseudoginsengenin DQ (PDQ) on cisplatin (CDDP)-induced nephrotoxicity were assessed, with a primary investigation into the mechanisms involved. Our results showed that pretreatment with PDQ remarkably restored levels of blood urea nitrogen (BUN) and creatinine (CRE), malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), tumor necrosis factor-α (TNF-α), and interleukin-1ß (IL-1ß). Meanwhile, PDQ decreased the CDDP-induced overexpression of heme oxygenase 1 (HO-1), cytochrome P450 E1 (CYP2E1), TNF-α, nuclear factor-kappa B (NF-κB), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) in renal tissues. Hoechst 33258 and TdT-mediated dUTP nick-end labeling (TUNEL) staining showed that CDDP-induced renal tubular cell apoptosis was apparently inhibited by PDQ. Western blotting showed that PDQ reversed the CDDP-induced (1) downregulation of Sirtuin-1 (Sirt-1), nuclear-related factor 2 (Nrf2), and Bcl-2, and (2) upregulation of NF-κB, Nox-4, Bax, caspase-9, and caspase-3. In addition, PDQ enhanced the antitumor activity of cisplatin in Lewis lung cancer xenograft tumor model mice. In conclusion, we found that PDQ exerted a renal protective effect against CDDP-induced acute nephrotoxicity via Sirt1/NF-κB and the caspase signaling pathway without compromising the antitumor activity of CDDP, which provides a new potential strategy for the clinical treatment of cancer and presents a new medicinal application of PDQ.


Assuntos
Injúria Renal Aguda/prevenção & controle , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Cisplatino/efeitos adversos , Triterpenos/administração & dosagem , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Animais , Apoptose/efeitos dos fármacos , Nitrogênio da Ureia Sanguínea , Cisplatino/administração & dosagem , Creatinina/metabolismo , Citocinas/genética , Modelos Animais de Doenças , Regulação para Baixo , Sinergismo Farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , Masculino , Malondialdeído/metabolismo , Camundongos , Triterpenos/química , Triterpenos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Mass Spectrom ; 52(10): 643-656, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28686313

RESUMO

Platycodon grandiflorum (PG) is extensively used for treating cough, excessive phlegm, sore throat, bronchitis and asthma, whereas Adenophora stricta (AS) is commonly used to reduce phlegm, clear lung and tonify stomach. Due to similar appearances, PG is sometimes adulterated with cheap AS so as to gain profits. And this will inevitably result in different pharmacological property. In order to further clarify the differences in the chemical composition of these two Chinese herbs, the ultra-high performance liquid chromatography combined with quadrupole time-of-flight tandem mass spectrometry coupled with UNIFI platform was used to establish a reliable, simple, sensitive and rapid analytical method. Seventy-five compounds, including triterpenoid saponins, organic acids, flavonoids, steroids, phenols, etc., were identified from PG based on MSE data and retention time under the optimized conditions. Meanwhile, 57 compounds including triterpenoid saponins, organic acids, steroids, phenols, alkaloids, etc. were identified from AS. Among all the identified compounds, there were only 14 common components (mainly organic acids) existing in two herbs, and most of the other chemical compositions are totally different between the two herbs. Based on the results, AS cannot substitute for PG. In addition, PG adulterated with AS will lead a poor efficacy in clinical application. In addition, the systematic comparison of similarities and differences between two Chinese herbs will provide reliable characterization profiles to clarify the pharmacological fundamental substances.


Assuntos
Campanulaceae/química , Medicamentos de Ervas Chinesas/análise , Platycodon/química , Cromatografia Líquida de Alta Pressão/métodos , Bases de Dados de Produtos Farmacêuticos , Contaminação de Medicamentos , Medicamentos de Ervas Chinesas/química , Humanos , Controle de Qualidade , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
16.
J Neurooncol ; 126(1): 19-26, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26476730

RESUMO

Tripartite motif (TRIM) proteins are involved in tumorigenesis. Here, we examined the expression, biological function, and clinical significance of tripartite motif containing 28 (TRIM28) in glioma, a locally aggressive brain tumor. First, TRIM28 expression was significantly higher in glioma (n = 138) than in non-glioma controls (n = 6). TRIM28 expression was positively correlated with tumor malignancy, and associated with poor overall survival (OS) and progression-free survival (PFS). Notably, TRIM28 expression was negatively correlated with p21 expression in patients with glioblastoma multiforme (GBM). A multivariate analysis that included relevant measures indicated that high TRIM28 expression is an independent prognostic factor for poor OS and PFS in GBM patients. In experiments with cultured glioma cells, down-regulating TRIM28 with shRNA increased p21 expression, and induced cell cycle arrest at the G1 phase. In a xenograft model, down-regulating TRIM28 suppressed tumor growth. These results indicate that over-expression of TRIM28 is associated with poor outcome in glioma patients.


Assuntos
Neoplasias Encefálicas/diagnóstico , Regulação da Expressão Gênica/genética , Glioma/diagnóstico , Proteínas Repressoras/metabolismo , Adulto , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/mortalidade , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Progressão da Doença , Feminino , Glioma/genética , Glioma/mortalidade , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/genética , Análise de Sobrevida , Fatores de Tempo , Proteína 28 com Motivo Tripartido , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Oncol Rep ; 35(1): 284-90, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26530859

RESUMO

miR-124 and Capn4 are aberrantly expressed in glioblastoma multiforme (GBM) tissues. In the present study, we investigated miR-124 and Capn4 expression in GBM tissue specimens. The role of miR-124 and Capn4 in the migration and invasion of glioma cells in vitro was also examined. miR-124 and Capn4 expression in 20 GBM and 6 control brain specimens was examined using RT-qPCR and immuno-blotting. Data from The Cancer Genome Atlas were retrieved. Candidate mRNA target sites of miR-124 were predicted using TargetScan/microRNA and binding was examined using dual luciferase reporter assays. The U87 and U251 cells were transfected with scrambled microRNA, miR-124 mimics and/or pLenti-Capn4 prior to wound­healing and Transwell invasion assays. Proteins involved in the epithelial-mesenchymal transition were examined using immunoblotting. The results showed that miR-124 was significantly downregulated in GBM tissues. Immunoblotting showed a marked upregulation of Capn4 expression in GBM tissues. The Spearman's correlation analysis revealed a negative association between miR-124 expression and Capn4 protein levels. TargetScan/microRNA predicted the miR-124 binding site in the nucleotide 440-446 region within the Capn4 3'-UTR, which was confirmed by luciferase assays. Wound­healing and Transwell invasion assays demonstrated that Capn4 downregulation or miR-124 mimics suppressed the migration and invasion of glioma cells. Capn4 downregulation or miR-124 mimics reduced the level of phospho-FAK and MMP2, vimentin and N-cadherin in U87 cells. In conclusion, miR-124 was found to suppress the migration and invasion of glioma cells in vitro via Capn4.


Assuntos
Neoplasias Encefálicas/patologia , Calpaína/genética , Glioblastoma/patologia , MicroRNAs/genética , Regiões 3' não Traduzidas , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Calpaína/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Técnicas In Vitro , MicroRNAs/metabolismo , Invasividade Neoplásica
18.
CNS Neurosci Ther ; 20(6): 521-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24628706

RESUMO

AIMS: Recent evidence indicates that the increased expression of calpain small subunit 1 (Capn4) is associated with tumorigenesis. This study was designed to explore the role which Capn4 plays in human glioma. METHODS: We detected the expression of Capn4 by immunohistochemistry in tissue microarrays and tissue samples. Following the down-regulation of Capn4 in glioma cell lines by a specific short hairpin RNA, the function of Capn4 in invasion, migration, and proliferation was assessed. We then evaluated the prognostic role of Capn4 using univariate and multivariate analysis in 94 glioblastoma (GBM) patients. RESULTS: Glioma tissues exhibited notably higher expression of Capn4 compared with control brain tissues and was positively correlated with histological malignancy. The down-regulation of Capn4 in glioma cells led to a decrease in invasion and migration in vitro. Through univariate analysis, the prognosis of GBM patients with Capn4 overexpression was significantly poorer with respect to progression-free survival (PFS) and overall survival (OS). Based on the results of the multivariate analysis, Capn4(high) was demonstrated to be a negative independent prognostic indicator for PFS and OS in GBM patients. CONCLUSION: The overexpression of Capn4 is a novel negative prognostic marker, and Capn4 may be used as a new target in therapeutic strategies for human glioma.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Calpaína/metabolismo , Glioma/metabolismo , Glioma/patologia , Adolescente , Adulto , Idoso , Calpaína/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Índice de Gravidade de Doença , Estatísticas não Paramétricas , Análise Serial de Tecidos , Cicatrização , Adulto Jovem
19.
Fish Shellfish Immunol ; 35(5): 1466-76, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24012751

RESUMO

The tumor suppressor protein p53 is a critical component of cell cycle checkpoint responses. It upregulates the expression of cyclin-dependent kinase inhibitors in response to DNA damage and other cellular perturbations, and promotes apoptosis when DNA repair pathways are overwhelmed. In the present study, the cDNA of p53 from the orange-spotted grouper (Epinephelus coioides) (Ec-p53) was cloned by the combination of homology cloning and rapid amplification of cDNA ends (RACE) approaches. The full-length cDNA of Ec-p53 was of 1921 bp, including an open reading frame (ORF) of 1143 bp encoding a polypeptide of 380 amino acids with predicted molecular weight of 42.3 kDa and theoretical isoelectric point of 7.0. Quantitative real-time PCR (qRT-PCR) assays revealed that Ec-p53 was ubiquitously expressed in all the examined tissues but with high levels in intestine and liver of the orange-spotted grouper. In addition, we measured the DNA damage and apoptosis in the blood cells and the percentage of dead and damaged blood cells. Our results suggest that oxidative stress and DNA damage occurred in grouper in conditions where the temperature was 15 ± 0.5 °C. Furthermore, qRT-PCR and western blot confirmed that low temperature stress induced upregulation of Ec-p53 in the mRNA and protein levels. These results suggest that low temperature-induced oxidative stress may cause DNA damage or apoptosis, and cooperatively stimulate the expression of Ec-p53, which plays a critical role in immune defense and antioxidant responses.


Assuntos
Regulação da Expressão Gênica/fisiologia , Perciformes/genética , Estresse Fisiológico/fisiologia , Temperatura , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Análise de Variância , Animais , Apoptose/fisiologia , Sequência de Bases , Western Blotting/veterinária , Clonagem Molecular , Ensaio Cometa/veterinária , Primers do DNA/genética , DNA Complementar/genética , Escherichia coli , Regulação da Expressão Gênica/genética , Hemócitos/metabolismo , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sequência de DNA/veterinária
20.
Traffic ; 8(9): 1164-78, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17605760

RESUMO

The members of the RGK small GTP-binding protein family, Kir/Gem, Rad, Rem and Rem2, are multifunctional proteins that regulate voltage-gated calcium channel activity and cell shape remodeling. Calmodulin (CaM) or CaM 14-3-3 are regulators of RGK functions and their association defines the subcellular localization of RGK proteins. Abolition of CaM association results in the accumulation of RGK proteins in the nucleus, whereas 14-3-3 binding maintains them in the cytoplasm. Kir/Gem possesses nuclear localization signals (NLS) that mediate nuclear accumulation through an importin alpha5-dependent pathway (see Mahalakshmi RN, Nagashima K, Ng MY, Inagaki N, Hunziker W, Béguin P. Nuclear transport of Kir/Gem requires specific signals and importin alpha5 and is regulated by Calmodulin and predicted service phosphorylations. Traffic 2007; doi: 10.1111/j.1600-0854.2007.00598.x). Because the extent of nuclear localization depends on the RGK protein and the cell type, the mechanism and regulation of nuclear transport may differ. Here, we extend our analysis to the other RGK members and show that Rem also binds importin alpha5, whereas Rad associates with importins alpha3, alpha5 and beta through three conserved NLS. Predicted phosphorylation of a serine residue within the bipartite NLS affects, as observed for Kir/Gem, nuclear accumulation of Rem, but not that of Rad or Rem2. We also identify an additional regulatory phosphorylation for all RGK proteins that prevents binding of 14-3-3 and thereby interferes with their cytosolic relocalization by 14-3-3. Functionally, nuclear localization of RGK proteins contributes to the suppression of RGK-mediated cell shape remodeling. Importantly, we show that endogenous RGK proteins are localized predominantly in the nucleus of individual cells of the brain cortex 'in situ' as well as in primary hippocampal cells, indicating that transport between the nucleus and their site of action in the cytoplasm (i.e., cytoskeleton, endoplasmic reticulum or plasma membrane) is of physiological relevance for the regulation of RGK protein function.


Assuntos
Núcleo Celular/metabolismo , Forma Celular/fisiologia , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Proteínas ras/fisiologia , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Células COS , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Células Cultivadas , Ceramidas/farmacologia , Chlorocebus aethiops , Células HeLa , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Camundongos , Modelos Biológicos , Proteínas Monoméricas de Ligação ao GTP/genética , Células NIH 3T3 , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/fisiologia , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Ratos , Proteínas ras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA