Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
2.
Clin Cosmet Investig Dermatol ; 16: 565-580, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36919011

RESUMO

Introduction: Secondary to war wounds, trauma, etc., hypertrophic scar formation is the cause of an excessive proliferation of fibroblasts and accumulation of collagen fibers, which might affect cosmetic appearance, and could cause malignant transformation. miRNAs play an important role in disease regulation via inhibiting post-transcriptional protein translation by targeting and binding to the 3' UTR region of mRNA. Here we explore the mechanism and interventions of scar formation from the perspective of miRNA. Methods: Hypertrophic scar-associated differential miRNAs were screened by analyzing sequencing data of normal skin and hypertrophic scar, and verified by RT-qPCR. Signaling pathways that may be influenced by differentially miRNAs were analyzed using KEGG and GO. miRNA mimics were used to explore the effects of miRNAs on SMAD signaling pathway proteins. Dual-luciferase assays were used to explore the targeted binding of miRNAs. The mimics of the miRNA were used to explore the impact of miRNAs on the proliferation, migration, apoptosis and collagen synthesis levels of hypertrophic scar fibroblasts. The scar model of rabbit ear was used to verify the influence of miRNA on wound healing and scar formation in vivo. Results: Expression of miR-182-5p was found to be considerably decreased in hypertrophic scars and fibroblasts. miR-182-5p was found to act mainly by targeting the 3'UTR region of SMAD4, but not SMAD1 or SMAD3. miR-182-5p overexpression may drastically suppress the proliferation and migration of fibroblasts, accompanied by enhanced apoptosis and reduced collagen fiber synthesis. The overexpression of miR-182-5p in in vivo experiments could effectively inhibit hypertrophic scar formation without affecting the speed and quality of wound healing. Conclusion: miR-182-5p inhibits hypertrophic scar formation by decreasing the proliferation and migration of fibroblasts via SMAD4 pathway, and is expected to become a novel hypertrophic scar therapeutic target.

3.
Acta Pharm Sin B ; 12(9): 3513-3528, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36176900

RESUMO

Recent insights collectively suggest the important roles of lysyl oxidase (LysOX) in the pathological processes of several acute and chronic neurological diseases, but the molecular regulatory mechanisms remain elusive. Herein, we explore the regulatory role of LysOX in the seizure-induced ferroptotic cell death of neurons. Mechanistically, LysOX promotes ferroptosis-associated lipid peroxidation in neurons via activating extracellular regulated protein kinase (ERK)-dependent 5-lipoxygenase (Alox5) signaling. In addition, overexpression of LysOX via adeno-associated viral vector (AAV)-based gene transfer enhances ferroptosis sensitivity and aggravates seizure-induced hippocampal damage. Our studies show that pharmacological inhibition of LysOX with ß-aminopropionitrile (BAPN) significantly blocks seizure-induced ferroptosis and thereby alleviates neuronal damage, while the BAPN-associated cardiotoxicity and neurotoxicity could further be reduced through encapsulation with bioresponsive amorphous calcium carbonate-based nanocarriers. These findings unveil a previously unrecognized LysOX-ERK-Alox5 pathway for ferroptosis regulation during seizure-induced neuronal damage. Suppressing this pathway may yield therapeutic implications for restoring seizure-induced neuronal injury.

4.
Front Oncol ; 12: 877657, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35646712

RESUMO

Neural infiltration is a critical component of the tumor microenvironment; however, owing to technological limitations, its role in hepatocellular cancer remains obscure. Herein, we obtained the RNA-sequencing data of liver hepatocellular carcinoma (LIHC) from The Cancer Genome Atlas database and performed a series of bioinformatic analyses, including prognosis analysis, pathway enrichment, and immune analysis, using the R software packages, Consensus Cluster Plus and Limma. LIHC could be divided into two subtypes according to the expression of neural-related genes (NRGs); moreover, there are statistic differences in the prognosis, stage, and immune regulation between the two subtypes. The prognostic model showed that high expression of NRGs correlated with a poor survival prognosis (P<0.05). Further, CHRNE, GFRA2, GFRA3, and GRIN2D was significantly correlated with LIHC clinical prognosis, clinical stage, immune infiltration, immune response, and vital signaling pathways. There was nerve-cancer crosstalk in LIHC. A reclassification of LIHC based on NRG expression may prove beneficial to clinical practice. CHRNE, GFRA2, GFRA3, and GRIN2D may serve as potential biomarker for liver cancer prognosis or immune response.

5.
J Allergy Clin Immunol ; 149(2): 579-588, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34547368

RESUMO

BACKGROUND: The epithelium is increasingly recognized as a pathologic contributor to asthma and its phenotypes. Although delayed wound closure by asthmatic epithelial cells is consistently observed, underlying mechanisms remain poorly understood, partly due to difficulties in studying dynamic physiologic processes involving polarized multilayered cell systems. Although type-2 immunity has been suggested to play a role, the mechanisms by which repair is diminished are unclear. OBJECTIVES: This study sought to develop and utilize primary multilayered polarized epithelial cell systems, derived from patients with asthma, to evaluate cell migration in response to wounding under type-2 and untreated conditions. METHODS: A novel wounding device for multilayered polarized cells, along with time-lapse live cell/real-time confocal imaging were evaluated under IL-13 and untreated conditions. The influence of inhibition of 15 lipoxygenase (15LO1), a type-2 enzyme, on the process was also addressed. Cell migration patterns were analyzed by high-dimensional frequency modulated Möbius for statistical comparisons. RESULTS: IL-13 stimulation negatively impacts wound healing by altering the total speed, directionality, and acceleration of individual cells. Inhibition 15LO1 partially improved the wound repair through improving total speed. CONCLUSIONS: Migration abnormalities contributed to markedly slower wound closure of IL-13 treated cells, which was modestly reversed by 15LO1 inhibition, suggesting its potential as an asthma therapeutic target. These novel methodologies offer new ways to dynamically study cell movements and identify contributing pathologic processes.


Assuntos
Asma/etiologia , Araquidonato 15-Lipoxigenase/fisiologia , Asma/diagnóstico por imagem , Asma/tratamento farmacológico , Asma/imunologia , Movimento Celular , Células Cultivadas , Células Epiteliais/fisiologia , Humanos , Interleucina-13/farmacologia , Inibidores de Lipoxigenase/farmacologia , Cicatrização/efeitos dos fármacos
6.
Mol Med Rep ; 25(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34751412

RESUMO

Vitiligo is a depigmentation disease commonly seen in clinical practice, mainly involving loss of functional epidermal pigment cells and hair follicle melanocytes. Narrow­band ultraviolet B (NB­UVB) has emerged as the first choice of treatment for vitiligo, but long­term exposure may have serious consequences. Recently, it was reported that adipose­derived stem cells (ADSCs) improve melanocyte growth and the efficacy of melanocyte transplantation. The present study aimed to examine the efficacy of NB­UVB/ADSC­transplantation combined therapy on a mouse vitiligo model and explore the underlying mechanisms by focusing on endoplasmic reticulum stress and cellular calcium (Ca2+) homeostasis. Vitiligo mice models were established by applying 40% monobenzone (MBZ) cream twice daily and treated with NB­UVB/ADSC combination therapy. Some treated mice were also given ML385, a nuclear factor erythroid 2 like 2 (Nr2) inhibitor. Histopathological changes were evaluated using a depigmentation evaluation score and observed with hematoxylin and eosin staining on skin tissues. ELISA was used to measure diagnostic markers in plasma. Flow cytometric assay was performed to quantify CD3+, CD4+ and CD8+ levels. Expression levels of associated proteins were detected with western blot and immunofluorescence. Treatment of mice with MBZ­induced depigmentation patches on the skin was accompanied with loss of redox balance and disruption of cellular Ca2+ homeostasis. Oxidative stress and Ca2+ unbalancing were improved after the mice were treated by NB­UVB/ADSCs transplantation combination therapy. ML385, strongly negated the protective effect of NB­UVB/ADSC transplantation combination therapy, indicating the critical role of Nr2 signaling. The findings improved the understanding of the pathogenesis of vitiligo and will guide future development of therapeutic strategies against it.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Pigmentação da Pele/fisiologia , Vitiligo/terapia , Animais , Cálcio/metabolismo , China , Estresse do Retículo Endoplasmático/fisiologia , Epiderme/metabolismo , Feminino , Folículo Piloso/metabolismo , Homeostase , Hidroquinonas/efeitos adversos , Hidroquinonas/farmacologia , Melanócitos/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/fisiologia , Estresse Oxidativo , Pele/patologia , Pigmentação da Pele/genética , Raios Ultravioleta , Terapia Ultravioleta/métodos , Vitiligo/metabolismo , Vitiligo/fisiopatologia
7.
Front Oncol ; 11: 753393, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34888238

RESUMO

BACKGROUND: Phosphorylated CTD-interacting factor 1 (PCIF1) is identified as the only known methyltransferase of N6,2'-O-dimethyladenosine (m6Am) in mRNA. However, its oncogenic and immunogenic role in cancer research is at an initial stage. METHODS: Herein, we carried out a pan-cancer analysis of PCIF1, with a series of datasets (e.g., TIMER2.0, GEPIA2, cBioPortal). RESULTS: PCIF1 expression was higher in most cancers than normal tissues and was discrepant across pathological stages. Highly expressed PCIF1 was positively correlated with overall survival (OS) or disease-free survival (DFS) of some tumors. PCIF1 expression had a positive correlation with CD4+ T-cell infiltration in kidney renal clear cell carcinoma (KIRC), CD8+ T cells, macrophages, and B cells in thyroid carcinoma (THCA), and immune checkpoint genes (ICGs) in LIHC but a negative correlation with CD4+ T cells, neutrophils, myeloid dendritic cells, and ICGs in THCA. It also affected tumor mutational burden (TMB) and microsatellite instability (MSI) of most tumors. CONCLUSION: PCIF1 expression was correlated with cancer prognosis and immune infiltration, suggesting it to be a potential target for cancer therapy.

8.
Front Immunol ; 12: 697964, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34135914

RESUMO

Cancer immunotherapy has achieved great advancement in the past decades. Whereas, its response is largely limited in immunologically cold tumors, in an urgent need to be solve. In recent years, an increasing number of studies have shown that inducing immunogenic cell deaths (ICDs) is an attractive approach to activate antitumor immunity. Upon specific stress, cancer cells undergo ICDs and dying cancer cells release danger associated molecular patterns (DAMPs), produce neoantigens and trigger adaptive immunity. ICDs exert a cancer vaccine-like effect and Inducement of ICDs mimics process of cancer vaccination. In this review, we propose a concept of ICD-based cancer vaccines and summarize sources of ICD-based cancer vaccines and their challenges, which may broaden the understandings of ICD and cancer vaccines in cancer immunotherapy.


Assuntos
Alarminas/imunologia , Vacinas Anticâncer/imunologia , Morte Celular Imunogênica/imunologia , Neoplasias/imunologia , Animais , Humanos
9.
Biochem Pharmacol ; 186: 114487, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33647264

RESUMO

Cancer immunotherapy (CIT) that targets the tumor immune microenvironment is regarded as a revolutionary advancement in the fight against cancer. The success and failure of CIT are due to the complexity of the immunosuppressive microenvironment. Cancer nanomedicine is a potential adjuvant therapeutic strategy for immune-based combination therapy. Exosomes are natural nanomaterials that play a pivotal role in mediating intercellular communications and package delivery in the tumor microenvironment. They affect the immune response or the effectiveness of immunotherapy. In particular, exosomal PD-L1 promotes cancer progression and resistance to immunotherapy. Exosomes possess high bioavailability, biological stability, targeting specificity, low toxicity, and immune characteristics, which indicate their potential for cancer therapy. They can be engineered to act as effective cancer therapeutic tools that activate anti-tumor immune response and start immune surveillance. In the current review, we introduce the role of exosomes in a tumor immune microenvironment, highlight the application of engineered exosomes to CIT, and discuss the challenges and prospects for clinical application.


Assuntos
Engenharia Química/métodos , Exossomos/imunologia , Imunoterapia/métodos , Nanopartículas/administração & dosagem , Neoplasias/imunologia , Neoplasias/terapia , Animais , Exossomos/metabolismo , Humanos , Nanopartículas/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/fisiologia
11.
Biomed Res Int ; 2020: 7134719, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204710

RESUMO

Adipose-derived stem cells (ADSCs) are an abundant cell source and provide an easy way to harvest mesenchymal stem cells, which are the focus of considerable attention in regenerative medicine. Electric fields (EF) play roles in many biological events and have been reported to promote cell proliferation, migration, and differentiation. In this study, ADSCs were treated with a direct current electric field (DCEF) of either 0 (control group) or 300 mV/mm (EF group) for six hours. RNA screening and analysis revealed that 66, 164, 26, and 1310 circRNAs, lncRNAs, miRNAs, and mRNAs, respectively, were differentially expressed in the DCEF-treated ADSCs compared with untreated ADSCs. Differentially expressed mRNAs were enriched in the MAPK signaling pathway, TNF signaling pathway, and some other pathways. ANXA1, ERRFI1, JAG1, EPHA2, PRR9, and H2AFY2 were related to the keratinocyte differentiation process. Competing endogenous RNA (ceRNA) networks were constructed on the basis of genes in the MAPK signaling pathway. Twenty-one RNAs in the above networks were randomly chosen, and their expression was validated using qRT-PCR, which showed the same expression trends as the RNA sequencing analysis. The MAPK signaling pathway is of great importance in the ADSC processes that occur in a DCEF, including keratinocyte differentiation. Several ceRNAs may participate in the regulation of MAPK signaling. This study may give new insight into the proliferation, migration, and differentiation of ADSCs, which will be valuable for tissue engineering and regenerative medicine.


Assuntos
Redes Reguladoras de Genes/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Células-Tronco Mesenquimais/fisiologia , Anexina A1/genética , Anexina A1/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Estimulação Elétrica , Regulação da Expressão Gênica , Ontologia Genética , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , RNA Circular/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Reprodutibilidade dos Testes
12.
Am J Cancer Res ; 10(8): 2242-2257, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32905502

RESUMO

The high mortality and poor clinical prognosis of glioblastoma multiforme (GBM) are concerns for many GBM patients as well as clinicians and researchers. The lack of a preclinical model that can easily be established and accurately recapitulate tumour biology and the tumour microenvironment further complicates GBM research and its clinical translation. GBM organoids (GBOs) are promising high-fidelity models that can be applied to model the disease, develop drugs, establish a living biobank, mimic therapeutic responses and explore personalized therapy. However, GBO models face some challenges, including deficient immune responses, absent vascular system and controversial reliability. In recent years, considerable progress has been achieved in the improvement of brain tumour organoid models and research based on such models. In addition to the traditional cultivation method, these models can be cultivated via genetic engineering and co-culture of cerebral organoids and GBM. In this review, we summarize the applications of GBM organoids and related advances and provide our opinions on associated limitations and challenges.

13.
Signal Transduct Target Ther ; 5(1): 166, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843638

RESUMO

Accumulating evidence shows that cellular and acellular components in tumor microenvironment (TME) can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Cancer research and treatment have switched from a cancer-centric model to a TME-centric one, considering the increasing significance of TME in cancer biology. Nonetheless, the clinical efficacy of therapeutic strategies targeting TME, especially the specific cells or pathways of TME, remains unsatisfactory. Classifying the chemopathological characteristics of TME and crosstalk among one another can greatly benefit further studies exploring effective treating methods. Herein, we present an updated image of TME with emphasis on hypoxic niche, immune microenvironment, metabolism microenvironment, acidic niche, innervated niche, and mechanical microenvironment. We then summarize conventional drugs including aspirin, celecoxib, ß-adrenergic antagonist, metformin, and statin in new antitumor application. These drugs are considered as viable candidates for combination therapy due to their antitumor activity and extensive use in clinical practice. We also provide our outlook on directions and potential applications of TME theory. This review depicts a comprehensive and vivid landscape of TME from biology to treatment.


Assuntos
Metformina/uso terapêutico , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Microambiente Tumoral/genética , Reposicionamento de Medicamentos , Humanos , Neoplasias/genética , Microambiente Tumoral/efeitos dos fármacos
14.
Trends Cancer ; 6(4): 265-267, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32209439

RESUMO

Patient-derived organoids can recapitulate parental tumor heterogeneity. In a recent study in Cell, Jacob et al. cultivated glioblastoma organoids (GBOs) to mimic tumor heterogeneity and chimeric antigen receptor (CAR)-T cell immunotherapy, applied it for xenograft establishment and drug testing, and generated a biobank for the timely start of post-operation therapy.


Assuntos
Glioblastoma , Receptores de Antígenos Quiméricos , Humanos , Organoides , Medicina de Precisão , Linfócitos T
15.
Chem Biol Interact ; 316: 108934, 2020 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-31870840

RESUMO

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most common lethal tumors with a high recurrence rate and low survival rate. Therefore, an urgent need exists for novel and effective treatment strategies for HNSCC patients. METHODS: Osthole, a natural ingredient extracted from Cnidium monnieri (L.) 'Cusson', has multiple pharmacological effects including antineoplastic activity. Regrettably, the antineoplastic effect of osthole in HNSCC cells remains undefined. We utilize in vitro assays to assess the anti-proliferative effects of osthole in HNSCC cells and tumorigenesis assays using FaDu cells in murine HNSCC models to assess in vivo function. Moreover, the possible molecular mechanisms of Osthole on HNSCC cells was also investigated. RESULTS: Our findings show that the anti-proliferation effect of osthole might function through induction of cell cycle arrest (G2/M phase) and apoptosis in HNSCC. Osthole could also down-regulating the protein level of cell cycle and apoptosis related proteins, such as Bcl-2, PARP1, Survivin, CyclinB1 and Cdc2, while up-regulating expression of Cleaved Caspase3/9, Cleaved PARP1 and Bax. Similarly, osthole suppressed the in vivo growth of FaDu cells in a subcutaneous tumor model. In terms of mechanism, our data show that osthole can suppress the PI3K/AKT pathway. CONCLUSIONS: In the current study, our in vitro and in vivo assay showed the suppressive effect of Osthole on HNSCC cells through induce cell cycle arrest (G2/M phase) and apoptosis. Moreover, the action mechanisms of Osthole on proliferation related signaling pathways was disclosed. Our present study suggests that osthole might be used as an effective therapeutic agent for patients with HNSCC.


Assuntos
Apoptose/efeitos dos fármacos , Cumarínicos/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Cnidium/química , Cnidium/metabolismo , Cumarínicos/química , Cumarínicos/uso terapêutico , Ciclina B1/genética , Ciclina B1/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia
16.
Front Oncol ; 9: 1225, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31799189

RESUMO

Long noncoding RNAs (lncRNAs) are emerging as important regulators of numerous biological processes, especially in cancer development. Aberrantly expressed and specifically located in tumor cells, they exert distinct functions in different cancers via regulating multiple downstream targets such as chromatins, RNAs, and proteins. Differentiation antagonizing non-protein coding RNA (DANCR) is a cytoplasmic lncRNA that generally works as a tumor promoter. Mechanically, DANCR promotes the functions of vital components in the oncogene network by sponging their corresponding microRNAs or by interacting with various regulating proteins. DANCR's distinct expression in tumor cells and collective involvement in pro-tumor pathways make it a promising therapeutic target for broad cancer treatment. Herein, we summarize the functions and molecular mechanism of DANCR in human cancers. Furthermore, we introduce the use of CRISPR/Cas9, antisense oligonucleotides and small interfering RNAs as well as viral, lipid, or exosomal vectors for onco-lncRNA targeted treatment. Conclusively, DANCR is a considerable promoter of cancers with a bright prospect in targeted therapy.

17.
JCI Insight ; 4(5)2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30730306

RESUMO

Although type-2-induced (T2-induced) epithelial dysfunction is likely to profoundly alter epithelial differentiation and repair in asthma, the mechanisms for these effects are poorly understood. A role for specific mucins, heavily N-glycosylated epithelial glycoproteins, in orchestrating epithelial cell fate in response to T2 stimuli has not previously been investigated. Levels of a sialylated MUC4ß isoform were found to be increased in airway specimens from asthmatic patients in association with T2 inflammation. We hypothesized that IL-13 would increase sialylation of MUC4ß, thereby altering its function and that the ß-galactoside α-2,6-sialyltransferase 1 (ST6GAL1) would regulate the sialylation. Using human biologic specimens and cultured primary human airway epithelial cells (HAECs),we demonstrated that IL-13 increases ST6GAL1-mediated sialylation of MUC4ß and that both were increased in asthma, particularly in sputum supernatant and/or fresh isolated HAECs with elevated T2 biomarkers. ST6GAL1-induced sialylation of MUC4ß altered its lectin binding and secretion. Both ST6GAL1 and MUC4ß inhibited epithelial cell proliferation while promoting goblet cell differentiation. These in vivo and in vitro data provide strong evidence for a critical role for ST6GAL1-induced sialylation of MUC4ß in epithelial dysfunction associated with T2-high asthma, thereby identifying specific sialylation pathways as potential targets in asthma.


Assuntos
Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Inflamação/metabolismo , Mucina-4/metabolismo , Sialiltransferases/metabolismo , Células Th2/imunologia , Adolescente , Adulto , Idoso , Antígenos CD/genética , Antígenos CD/farmacologia , Asma/imunologia , Linhagem Celular , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Interleucina-13 , Pulmão , Masculino , Pessoa de Meia-Idade , Mucina-4/genética , Isoformas de Proteínas , Sialiltransferases/genética , Sialiltransferases/farmacologia , Células Th2/efeitos dos fármacos , Transcriptoma , Adulto Jovem
18.
Front Oncol ; 8: 598, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30581774

RESUMO

Chemotherapy with or without radiation remains the first choice for most cancers. However, intolerant side effects and conventional drug resistance restrict actual clinical efficacy. Curaxin CBL0137 is designed to regulate p53 and nuclear factor-κB simultaneously and to prevent the resistance caused by a single target. Functionally, CBL0137 exhibits an antitumor activity in multiple cancers, including glioblastoma, renal cell carcinoma, melanoma, neuroblastoma, and small cell lung cancer (SCLC). Mechanistically, CBL0137 is originally identified to act by facilitates chromatin transcription (FACT) complex. Further investigations reveal that several pathways, such as NOTCH1 and heat shock factor 1 (HSF1), are involved in the process. CBL0137 has been reported to target cancer stem cells (CSCs) and enhance chemotherapy/monotherapy efficacy. The translational advance of CBL0137 into clinical practice is expected to provide a promising future for cancer treatment.

19.
Cancer Sci ; 109(7): 2199-2210, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29788550

RESUMO

Ubiquitin-specific protease 22 (USP22) is a member of the "death-from-cancer" signature, which plays a key role in cancer progression. Previous evidence has shown that USP22 is overexpressed and correlates with poor prognosis in glioma. The effect and mechanism of USP22 in glioma malignancy, especially cancer stemness, remain elusive. Herein, we find USP22 is more enriched in stem-like tumorspheres than differentiated glioma cells. USP22 knockdown inhibits cancer stemness in glioma cell lines. With a cell-penetrating TAT-tag protein, B cell-specific Moloney murine leukemia virus integration site 1 (BMI1), a robust glioma stem-cell marker, is found to mediate the effect of USP22 on glioma stemness. By immunofluorescence, USP22 and BMI1 are found to share similar intranuclear expression in glioma cells. By analysis with immunohistochemistry and bioinformatics, USP22 is found to positively correlate with BMI1 at the post-translational level only rather than at the transcriptional level. By immunoprecipitation and in vivo deubiquitination assay, USP22 is found to interact with and deubiquitinate BMI1 for protein stabilization. Microarray analysis shows that USP22 and BMI1 mutually regulate a series of genes involved in glioma stemness such as POSTN, HEY2, PDGFRA and ATF3. In vivo study with nude mice confirms the role of USP22 in promoting glioma tumorigenesis by regulating BMI1. All these findings indicate USP22 as a novel deubiquitinase of BMI1 in glioma. We propose a working model of the USP22-BMI1 axis, which promotes glioma stemness and tumorigenesis through oncogenic activation. Thus, targeting USP22 might be an effective strategy to treat glioma especially in those with elevated BMI1 expression.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Complexo Repressor Polycomb 1/metabolismo , Tioléster Hidrolases/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Transformação Celular Neoplásica/genética , Glioma/metabolismo , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Proteínas Oncogênicas , Ubiquitina Tiolesterase
20.
Cancer Lett ; 414: 174-180, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29174804

RESUMO

Cancer harbors variable heterogeneity and plasticity. Thus far, our comprehension is greatly based on cell lines, organoids, and patient-derived tumor xenografts (PDTXs). Organoids are a three-dimensional in vitro culture platform constructed from self-organizing stem cells. They can almost accurately recapitulate tumor heterogeneity and microenvironment "in a dish," which surpass established cell lines and are not as expensive and time-consuming as PDTXs. As an intermediate model, tumor organoids are also used to study the fundamental issues of tumorigenesis and metastasis. They are specifically applied for drug testing and stored as "living biobanks." In this review, we highlight the translational applications of organoid technologies in tumor research and precision medicine, discuss the advantages and limitations compared with other mentioned methods, and provide our outlook on its future.


Assuntos
Modelos Biológicos , Neoplasias/metabolismo , Organoides/metabolismo , Técnicas de Cultura de Tecidos/métodos , Microambiente Tumoral , Animais , Humanos , Neoplasias/genética , Neoplasias/patologia , Especificidade de Órgãos , Organoides/patologia , Medicina de Precisão/métodos , Pesquisa Translacional Biomédica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA