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










Intervalo de ano de publicação
1.
Front Pharmacol ; 15: 1420643, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38962310

RESUMO

Lung cancer, recognized globally as a leading cause of malignancy-associated morbidity and mortality, is marked by its high prevalence and lethality, garnering extensive attention within the medical community. Mitophagy is a critical cellular process that plays a crucial role in regulating metabolism and ensuring quality control within cells. Its relevance to lung cancer has garnered significant attention among researchers and scientists. Mitophagy's involvement in lung cancer encompasses its initiation, progression, metastatic dissemination and treatment. The regulatory landscape of mitophagy is complex, involving numerous signaling proteins and pathways that may exhibit aberrant alterations or mutations within the tumor environment. In the field of treatment, the regulation of mitophagy is considered key to determining cancer chemotherapy, radiation therapy, other treatment options, and drug resistance. Contemporary investigations are directed towards harnessing mitophagy modulators, both inhibitors and activators, in therapeutic strategies, with an emphasis on achieving specificity to minimize collateral damage to healthy cellular populations. Furthermore, molecular constituents and pathways affiliated with mitophagy, serving as potential biomarkers, offer promising avenues for enhancing diagnostic accuracy, prognostic assessment, and prediction of therapeutic responses in lung cancer. Future endeavors will also involve investigating the impact of mitophagy on the composition and function of immune cells within the tumor microenvironment, aiming to enhance our understanding of how mitophagy modulates the immune response to lung cancer. This review aims to comprehensively overview recent advancements about the role of mitophagy in the tumor genesis, progenesis and metastasis, and the impact of mitophagy on the treatment of lung cancer. We also discussed the future research direction of mitophagy in the field of lung cancer.

2.
Res Sq ; 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38405979

RESUMO

Background: Kindlin-2, an adaptor protein, is dysregulated in various human cancers, including triple negative breast cancer (TNBC), where it drives tumor progression and metastasis by influencing several cancer hallmarks. One well-established role of Kindlin-2 involves the regulation of integrin signaling, achieved by directly binding to the cytoplasmic tail of the integrin ß subunit. In this study, we present novel insights into Kindlin-2's involvement in stabilizing the ß1-Integrin:TGF-ß type 1 receptor (TßRI) complexes, acting as a physical bridge that links ß1-Integrin to TßRI. The loss of Kindlin-2 results in the degradation of this protein complex, leading to the inhibition of downstream oncogenic pathways. Methods: Our methodology encompassed a diverse range of in vitro assays, including CRISPR/Cas9 gene editing, cell migration, 3D tumorsphere formation and invasion, solid binding, co-immunoprecipitation, cell adhesion and spreading assays, as well as western blot and flow cytometry analyses, utilizing MDA-MB-231 and 4T1 TNBC cell lines. Additionally, preclinical in vivo mouse models of TNBC tumor progression and metastasis were employed to substantiate our findings. Results: The investigation revealed that the direct interaction between Kindlin-2 and ß1-Integrin is mediated through the C-terminal F3 domain of Kindlin-2, while the interaction between Kindlin-2 and TßRI is facilitated through the F2 domain of Kindlin-2. Disruption of this bridge, achieved via CRISPR/Cas9-mediated knockout of Kindlin-2, led to the degradation of ß1-Integrin and TßRI, resulting in the inhibition of oncogenic pathways downstream of both proteins, subsequently hindering tumor growth and metastasis. Treatment of Kindlin-2-deficient cells with the proteasome inhibitor MG-132 restored the expression of both ß1-Integrin and TßRI. Furthermore, the rescue of Kindlin-2 expression reinstated their oncogenic activities both in vitro and in vivo. Conclusions: This study identifies a novel function of Kindlin-2 in stabilizing the ß1-Integrin:TßR1 complexes and regulating their downstream oncogenic signaling. The translational implications of these findings are substantial, potentially unveiling new therapeutically targeted pathways crucial for the treatment of TNBC tumors.

3.
Front Pharmacol ; 14: 1243727, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38026959

RESUMO

Objective: Non-small cell lung cancer (NSCLC) is recognized for its aggressive nature and propensity for high rates of metastasis. The NLRP3 inflammasome pathway plays a vital role in the progression of NSCLC. This study aimed to investigate the effects of S. exigua extract and its active compounds on NLRP3 regulation in NSCLC using an in vitro model. Methods: S. exigua was extracted using hexane, ethyl acetate and ethanol to obtain S. exigua hexane fraction (SE-Hex), S. exigua ethyl acetate fraction (SE-EA), and S. exigua ethanol fraction (SE-EtOH) respectively. The active compounds were identified using column chromatography and NMR analysis. A549 cells were primed with lipopolysaccharide (LPS) and adenosine triphosphate (ATP) for activated NLRP3 inflammasome. The anti-inflammatory properties were determined using ELISA assay. The anti-proliferation and anti-metastasis properties against LPS-ATP-induced A549 cells were determined by colony formation, cell cycle, wound healing, and trans-well migration and invasion assays. The inflammatory gene expressions and molecular mechanism were determined using RT-qPCR and Western blot analysis, respectively. Results: SE-EA exhibited the greatest anti-inflammation properties compared with other two fractions as evidenced by the significant inhibition of IL-1ß, IL-18, and IL-6, cytokine productions from LPS-ATP-induced A549 cells in a dose-dependent manner (p < 0.05). The analysis of active compounds revealed exiguaflavanone A (EGF-A) and exiguaflavanone B (EGF-B) as the major compounds present in SE-EA. Then, SE-EA and its major compound were investigated for the anti-proliferation and anti-metastasis properties. It was found that SE-EA, EGF-A, and EGF-B could inhibit the proliferation of LPS-ATP-induced A549 cells through cell cycle arrest induction at the G0/G1 phase and reducing the expression of cell cycle regulator proteins. Furthermore, SE-EA and its major compounds dose-dependently suppressed migration and invasion of LPS-ATP-induced A549 cells. At the molecular level, SE-EA, EGF-A, and EGF-B significantly downregulated the mRNA expression of IL-1ß, IL-18, IL-6, and NLRP3 in LPS-ATP-induced A549 cells. Regarding the mechanistic study, SE-EA, EGF-A, and EGF-B inhibited NLRP3 inflammasome activation through suppressing NLRP3, ASC, pro-caspase-1(p50 form), and cleaved-caspase-1(p20 form) expressions. Conclusion: Targeting NLRP3 inflammasome pathway holds promise as a therapeutic approach to counteract pro-tumorigenic inflammation and develop novel treatments for NSCLC.

4.
Oral Dis ; 2023 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-37530513

RESUMO

OBJECTIVE: Microbial dysbiosis and microbiome-induced inflammation may play a role in the etiopathogenesis of oral squamous cell carcinoma (OSCC). Candida albicans (C. albicans) is the most prevalent opportunistic pathogenic fungus in the oral cavity, and Candida infection is considered as one of its high-risk factors. Although oral microbiota-host interactions are closely associated with the development of OSCC, the interrelationship between fungi and OSCC is poorly understood compared to that between bacteria and viruses. RESULTS: We accumulated knowledge of the evidence, pathogenic factors, and possible multiple mechanisms by which C. albicans promotes malignant transformation of OSCC, focusing on the induction of epithelial damage, production of carcinogens, and regulation of the tumor microenvironment. In addition, we highlight the latest treatment strategies for Candida infection. CONCLUSION: This review provides a new perspective on the interrelationship between C. albicans and OSCC and contributes to the establishment of a systematic and reliable clinical treatment system for OSCC patients with C. albicans infection.

5.
Front Biosci (Landmark Ed) ; 27(9): 263, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36224007

RESUMO

Cancer progression and metastases are the leading causes of poor outcomes in patients with colon cancer. Colon cancer metastasis is a multigene, multistep, multistage complex process in which target genes, microRNAs, epithelial-stromal transformation, tumour stem cells, the tumour microenvironment, and various cell signalling pathways are implicated in the progression and metastasis of colon cancer. Although conventional therapies have made significant advances in treating the progression and metastasis of colorectal cancer, they have failed to improve survival outcomes. Natural compounds may have more significant potential in preventing and treating colon cancer. Active natural compounds exert their antitumor effects by inducing tumour cell differentiation, promoting tumour cell apoptosis, inhibiting tumour vascular growth, and regulating immunity. Natural compounds, combined with conventional therapies, can target mutant genes and various cellular signalling pathways, inhibit epithelial-stromal transformation, and improve the tumour microenvironment to inhibit tumour progression and metastasis. The synergism of natural compounds and conventional therapeutics has the potential to become a promising therapy for treating colorectal cancer progression and metastases.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , MicroRNAs , Apoptose , Neoplasias do Colo/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Humanos , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral
6.
Breast Cancer Res ; 24(1): 48, 2022 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-35836253

RESUMO

BACKGROUND: A pro-oxidant enzyme, NADPH oxidase 4 (Nox4) has been reported to be a critical downstream effector of TGFß-induced myofibroblast transformation during fibrosis. While there are a small number of studies suggesting an oncogenic role of Nox4 derived from activated fibroblasts, direct evidence linking this pro-oxidant to the tumor-supporting CAF phenotype and the mechanisms involved are lacking, particularly in breast cancer. METHODS: We targeted Nox4 in breast patient-derived CAFs via siRNA-mediated knockdown or administration of a pharmaceutical inhibitor (GKT137831). We also determine primary tumor growth and metastasis of implanted tumor cells using a stable Nox4-/- syngeneic mouse model. Autophagic flux of CAFs was assessed using a tandem fluorescent-tagged ptfl-LC3 plasmid via confocal microscopy analysis and determination of the expression level of autophagy markers (beclin-1 and LC3B). Nox4 overexpressing CAFs depend on the Nrf2 (nuclear factor-erythroid factor 2-related factor 2) pathway for survival. We then determined the dependency of Nox4-overexpressing CAFs on the Nrf2-mediated adaptive stress response pathway for survival. Furthermore, we investigated the involvement of Birc5 on CAF phenotype (viability and collagen contraction activity) as well as the expression level of CAF markers, FAP and αSMA. CONCLUSIONS: We found that deletion of stroma Nox4 and pharmaceutically targeting its activity with GKT137831 significantly inhibited orthotopic tumor growth and metastasis of implanted E0771 and 4T1 murine mammary carcinoma cell lines in mice. More importantly, we found a significant upregulation of Nox4 expression in CAFs isolated from human breast tumors versus normal mammary fibroblasts (RMFs). Our in situ RNA hybridization analysis for Nox4 transcription on a human breast tumor microarray further support a role of this pro-oxidant in the stroma of breast carcinomas. In addition, we found that Nox4 promotes autophagy in CAFs. Moreover, we found that Nox4 promoted survival of CAFs via activation of Nrf2, a master regulator of oxidative stress response. We have further shown Birc5 is involved as a downstream modulator of Nrf2-mediated pro-survival phenotype. Together these studies indicate a role of redox signaling via the Nox4-Nrf2 pathway in tumorigenesis and metastasis of breast cancer cells by promoting autophagy and survival of CAFs.


Assuntos
Neoplasias da Mama , Fibroblastos Associados a Câncer , Animais , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/patologia , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Feminino , Fibroblastos/metabolismo , Humanos , Camundongos , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Survivina/metabolismo , Regulação para Cima
7.
Front Oncol ; 12: 857686, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35372069

RESUMO

The ability of cancer cells to adjust their metabolism in response to environmental changes is a well-recognized hallmark of cancer. Diverse cancer and non-cancer cells within tumors compete for metabolic resources. Metabolic demands change frequently during tumor initiation, progression and metastasis, challenging our quest to better understand tumor biology and develop novel therapeutics. Vascularization, physical constraints, immune responses and genetic instability promote tumor evolution resulting in immune evasion, opportunities to breach basement membrane barriers and spread through the circulation and lymphatics. In addition, the unfolded protein response linked to the ubiquitin proteasome system is a key player in addressing stoichiometric imbalances between nuclear and mitochondrially-encoded protein subunits of respiratory complexes, and nuclear-encoded mitochondrial ribosomal protein subunits. While progressive genetic changes, some of which affect metabolic adaptability, contribute to tumorigenesis and metastasis through clonal expansion, epigenetic changes are also important and more dynamic in nature. Understanding the role of stromal and immune cells in the tumor microenvironment in remodeling cancer cell energy metabolism has become an increasingly important area of research. In this perspective, we discuss the adaptations made by cancer cells to balance mitochondrial and glycolytic energy metabolism. We discuss how hypoxia and nutrient limitations affect reductive and oxidative stress through changes in mitochondrial electron transport activity. We propose that integrated responses to cellular stress in cancer cells are central to metabolic flexibility in general and bioenergetic adaptability in particular and are paramount in tumor progression and metastasis.

8.
Neoplasia ; 23(11): 1147-1165, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34706306

RESUMO

Epithelial-mesenchymal transition (EMT) contributes to tumor invasion, metastasis and drug resistance. AKT activation is key in a number of cellular processes. While many positive regulators for either EMT or AKT activation have been reported, few negative regulators are established. Through kinase cDNA screen, we identified brain-type creatine kinase (CKB or BCK) as a potent suppressor for both. As a ubiquitously expressed kinase in normal tissues, CKB is significantly downregulated in several solid cancer types. Lower CKB expression is significantly associated with worse prognosis. Phenotypically, CKB overexpression suppresses, while its silencing promotes, EMT and cell migration, xenograft tumor growth and metastasis of prostate cancer cells. AKT activation is one of the most prominent signaling events upon CKB silencing in prostate cancer cells, which is in line with prostate cancer TCGA data. EMT enhanced by CKB silencing is abolished by AKT inhibition. Mechanistically, CKB interacts with AKT and sequestrates it from activation by mTOR. We further elucidated that an 84aa fragment at C-terminus of CKB protein interacts with AKT's PH domain. Ectopic expression of the 84aa CKB fragment inhibits AKT activation, EMT and cell proliferation. Interestingly, molecular dynamics simulation on crystal structures of AKT and CKB independently demonstrates that AKT's PH domain and CKB's 84aa fragment establish their major interaction interface. In summary, we have discovered CKB as a negative regulator of EMT and AKT activation, revealing a new mode of their regulation . We have also demonstrated that CKB downregulation is a poor prognosticator, which is sufficient to promote prostate cancer progression.


Assuntos
Biomarcadores Tumorais/metabolismo , Creatina Quinase Forma BB/farmacologia , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Prognóstico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Endocr Relat Cancer ; 28(10): 657-670, 2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34310341

RESUMO

Steroid receptor coactivators (SRCs) possess specific and distinct oncogenic roles in the initiation of cancer and in its progression to a more aggressive disease. These coactivators interact with nuclear receptors and other transcription factors to boost transcription of multiple genes, which potentiate cancer cell proliferation, migration, invasion, tumor angiogenesis and epithelial-mesenchymal transition (EMT). Targeting SRCs using small molecule inhibitors (SMIs) is a promising approach to control cancer progression and metastasis. By high-throughput screening analysis, we recently identified SI-2 as a potent SRC SMI. To develop therapeutic agents, SI-10 and SI-12, the SI-2 analogs are synthesized that incorporate the addition of F atoms to the SI-2 chemical structure. As a result, these analogs exhibit a significantly prolonged plasma half-life, minimal toxicity and improved hERG activity. Biological functional analysis showed that SI-10 and SI-12 treatment (5-50 nM) can significantly inhibit viability, migration and invasion of breast cancer cells in vitro and repress the growth of breast cancer PDX organoids. Treatment of mice with 10 mg/kg/day of either SI-10 or SI-12 was sufficient to repress the growth of xenograft tumors derived from MDA-MB-231 and LM2 cells. Furthermore, in spontaneous and experimental metastasis mouse models developed from MDA-MB-231 and LM2 cells, respectively, SI-10 and SI-12 effectively inhibited the progression of breast cancer lung metastasis. These results demonstrate that SI-10 and SI-12 are promising therapeutic agents and are specifically effective in blocking tumor metastasis, a key point in tumor progression to a more lethal state that results in patient mortality in the majority of cases.


Assuntos
Neoplasias da Mama , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Humanos , Camundongos , Coativador 3 de Receptor Nuclear/antagonistas & inibidores , Oncogenes
10.
Int J Mol Sci ; 21(15)2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32751958

RESUMO

Hypoxia in locally advanced solid tumors develops due to uncontrollable cell proliferation, altered metabolism, and the severe structural and functional abnormality of the tumor vasculature, leading to an imbalance between oxygen supply and consumption in the fast-growing tumors and negative impact on the therapeutic outcome. Several hypoxia-responsive molecular determinants, such as hypoxia-inducible factors, guide the cellular adaptation to hypoxia by gene activation, which is critical for promoting malignant progression in the hostile tumor microenvironment. Over time, a large body of evidence exists to suggest that tumor hypoxia also influences the tumor metabolic reprogramming, resulting in neoangiogenesis, metastasis, and immune evasion. In this respect, our review aims to understand the biological processes, key events, and consequences regarding the hypoxia-driven metabolic adaptation of tumor cells. We also assess the potential therapeutic impact of hypoxia and highlight our review by discussing possible therapeutic strategies targeting hypoxia, which would advance the current understanding of hypoxia-associated tumor propagation and malignant progression and improve the management of tumor hypoxia.


Assuntos
Neoplasias/metabolismo , Hipóxia Tumoral , Proliferação de Células , Progressão da Doença , Humanos , Neovascularização Patológica , Microambiente Tumoral
11.
Adv Exp Med Biol ; 1258: 189-197, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32767243

RESUMO

It is becoming increasingly recognized that the tumor microenvironment significantly contributes to the development, progression, and metastasis of cancer and also plays a role in response to treatment. The tumor microenvironment is a complex and heterogeneous niche comprised of stromal cells, cancer cells, blood vessels, areas of hypoxia and necrotic tissue, fibrosis, and extracellular matrix. Cellular communication takes place within the tumor microenvironment, both via cell to cell contact, and through extracellular mechanisms such as exosomal signaling. Exosomes are very small membrane-bound vesicles that have been shown to play key roles in the progression of cancer including modulation of the tumor microenvironment through the induction of angiogenesis, the transfer of genetic information that confers drug resistance, and increased cell migration, invasion, proliferation, and survival, as well as the modulation of immune cell interactions. The role of exosomes in several different cancers has been investigated. In the context of osteosarcoma, understanding how exosomes may modulate the tumor microenvironment to support metastatic growth particularly in the lung, the most common site of metastases, may identify novel therapeutic targets for relapsed patients.


Assuntos
Comunicação Celular , Exossomos , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais , Microambiente Tumoral , Humanos
12.
J Cell Mol Med ; 23(3): 1873-1884, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30588744

RESUMO

Long non-coding RNAs (lncRNAs) play essential roles in diverse biological processes; however, current understanding of the mechanism underlying the regulation of tumour proliferation and metastasis is limited. Lung cancer-associated transcript 1 (LUCAT1) has been reported in a variety of human cancers, while its role in hepatocellular carcinoma (HCC) remains unclear. This study aimed to determine the biological role and underlying mechanism of LUCAT1 on progression and metastasis in HCC cells and clinical specimens. Our results demonstrated that LUCAT1 was up-regulated in HCC tissues and cells. Loss- and gain-of-function studies revealed that LUCAT1 promotes the proliferation and metastasis of HCC cells in vitro and in vivo. Furthermore, RNA pulldown and Western blot assays indicated that LUCAT1 inhibited the phosphorylation of Annexin A2 (ANXA2) to reduce the degradation of ANXA2-S100A10 heterotetramer (AIIt), which in turn accelerated the secretion of plasminogen into plasmin, thereby resulting in the activation of metalloprotease proteins. In conclusion, we propose that LUCAT1 serves as a novel diagnostic and therapeutic target for HCC.


Assuntos
Anexina A2/genética , Carcinogênese/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , RNA Longo não Codificante/genética , Biomarcadores Tumorais/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Células Hep G2 , Humanos , MicroRNAs/genética , Estadiamento de Neoplasias/métodos , Fosforilação/genética , Interferência de RNA/fisiologia , Regulação para Cima/genética
13.
Biochem Biophys Res Commun ; 509(1): 143-147, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30573358

RESUMO

Long non-coding RNA FENDRR is implicated in progression of several cancers, but its exact role and mechanism in hepatocellular carcinoma (HCC) are largely unknown. In this study, we investigated the expression and biological roles of FENDRR in HCC tissues and cell lines. We found that the expression levels of FENDRR were significantly down-regulated in HCC tissues and cells. FENDRR overexpression could inhibit the growth of HCC cells in vitro and in vivo. Moreover, up-regulation of FENDRR suppressed the migration and invasion of HCC cells. Mechanistically, we demonstrated that FENDRR interacted directly with Glypican-3 (GPC3) promoter and methylated GPC3 promoter, which led to down-regulation of GPC3 expression. Ectopic expression of GPC3 ablated the inhibitory effects of FENDRR on HCC cell proliferation, migration and invasion. Taken together, we provided the first evidence for the inhibitory activity of FENDRR in HCC, which is causally linked to targeting GPC3 at the epigenetic level. Restoration of FENDRR may be a potential approach to prevent HCC progression and metastasis.


Assuntos
Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Glipicanas/genética , Neoplasias Hepáticas/genética , RNA Longo não Codificante/genética , Adulto , Idoso , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Regulação para Baixo , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia
14.
J Craniomaxillofac Surg ; 46(10): 1748-1752, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30145047

RESUMO

PURPOSE: This study investigates the possibility and sensibility of using sentinel node biopsy (SNB) during surgery for oropharyngeal carcinomas with clinically and radiographically unremarkable cervical lymph nodes. MATERIALS AND METHODS: A total of 36 patients who were treated for early oral and oropharyngeal carcinoma and unremarkable cervical lymph nodes were included in this study. After lymphoscintigraphy for detecting sentinel lymph nodes (SLN), the SLN were excised first. Thereafter elective neck dissection was performed. Histopathological and immunochemical examinations were used to examine the SLN and all other lymph nodes. RESULTS: The preoperative SLN detection rate was 97.2% (35 of 36). SLN in level V were detected in four patients (11.1%). Metastases were found in 33.3% of the patients (12 of 36). All metastases were in the marked SLN. No skip metastases could be detected. A specificity of 100% and a sensitivity of 94.4% were identified for the SNB. CONCLUSION: The study confirms that SNB is a precise diagnostic procedure for assessing the nodal status of cervical lymph nodes. Further studies are needed to determine whether SNB without elective neck dissection for clinically and radiologically unremarkable cervical lymph nodes can become a reliable course of treatment for carcinomas of the head and neck region.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/patologia , Neoplasias Orofaríngeas/patologia , Biópsia de Linfonodo Sentinela , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/cirurgia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/cirurgia , Esvaziamento Cervical , Estadiamento de Neoplasias/métodos , Neoplasias Orofaríngeas/cirurgia , Reprodutibilidade dos Testes
15.
J Orthop Res ; 2018 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-29427436

RESUMO

Osteosarcoma is rare and little improvement in survival rates has occurred in the last 25 years despite modern chemotherapeutic treatment. Bioluminescent cell lines for the modeling of osteosarcoma have shown success in tracking metastases in vivo, but commonly use adenoviral vectors to transfect the native cell line with bioluminescent reporters. The purpose of this study was to develop an orthotopic model for metastatic osteosarcoma capable of in vivo monitoring of metastatic and primary tumor burden in an immunocompetent mouse and compare that model to its wild type pathogenesis. K7M2 cells were transfected using a plasmid vector and were stable after 12 weeks. Thirty-four female BALB/c mice aged 4-5 weeks underwent orthotopic implantation of either wild type (n = 12) or transfected (n = 22) K7M2 cells in the proximal tibia. Mice were monitored for tumor growth and weekly In Vivo Imaging System (IVIS) imaging was performed to monitor for pulmonary metastasis. Although tumors developed sooner in the wild type group, no significant differences were seen compared to Transfected Group 1 in rate of inoculation, growth rates after first detection, metastatic rate, and time between inoculation and death. This study establishes a new murine model for metastatic osteosarcoma using the K7M2-wt cell line transfected with a non-viral plasmid luciferase vector. The benefits of this preclinical model include an intact immune system and orthotopically driven metastatic disease; this model appears comparable to its wild type counterpart. In the future, the model may be used to examine promising immunomodulatory therapies using bioluminescence in vivo. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.

16.
J Cell Commun Signal ; 10(3): 241-249, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27473382

RESUMO

Exosomes, which act as biological cargo vessels, are cell-released, phospholipid-enclosed vesicles. In eukaryotic cells, exosomes carry and exchange biological materials or signals for the benefit or detriment to the cells. Thereby, we consider exosomes to be molecular Palkis (carriers). Although exosomes are currently one of the most popularly researched cellular entities, they have remained largely enigmatic and warrant continued investigation into their structure and functions. These membraned vesicles are between 30 and 150 nm in diameter and are actively secreted by all cell types. While initially considered cellular "trash bags," recent years have revealed exosomes to be dynamic and multi-functional vesicles that may play a crucial role in cancer development, progression and metastasis. Thereby, they have the potential to be used in development of therapeutic modalities for cancer and other diseases. As more research studies emerge, it's becoming evident that exosomes are released by cells with a purpose and are representatives of certain cell types and disease conditions. Hence, they may also be used as biomarkers for the detection of cancer initiation, progression and organotropic metastatic growth of cancer cells. This review will focus on the recent developments achieved in identifying the role of exosomes in cancer development and progression as well as therapeutic implications. The review will also discuss the pitfalls of methodologies used for the extraction of exosomes.

17.
J Biol Chem ; 291(31): 16068-81, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27261462

RESUMO

Loss of 14-3-3σ has been observed in multiple tumor types; however, the mechanisms by which 14-3-3σ loss leads to tumor progression are not understood. The experiments in this report demonstrate that loss of 14-3-3σ leads to a decrease in the expression of epithelial markers and an increase in the expression of mesenchymal markers, which is indicative of an induction of the epithelial to mesenchymal transition (EMT). The EMT was accompanied by an increase in migration and invasion in the 14-3-3σ(-/-) cells. 14-3-3σ(-/-) cells show increased stabilization of c-Jun, resulting in an increase in the expression of the EMT transcription factor slug. 14-3-3σ induces the ubiquitination and degradation of c-Jun in an FBW7-dependent manner. c-Jun ubiquitination is dependent on the presence of an intact nuclear export pathway as c-Jun is stabilized and localized to the nucleus in the presence of a nuclear export inhibitor. Furthermore, the absence of 14-3-3σ leads to the nuclear accumulation and stabilization of c-Jun, suggesting that 14-3-3σ regulates the subcellular localization of c-Jun. Our results have identified a novel mechanism by which 14-3-3σ maintains the epithelial phenotype by inhibiting EMT and suggest that this property of 14-3-3σ might contribute to its function as a tumor suppressor gene.


Assuntos
Proteínas 14-3-3/metabolismo , Núcleo Celular/metabolismo , Transição Epitelial-Mesenquimal , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação , Proteínas 14-3-3/genética , Transporte Ativo do Núcleo Celular/genética , Linhagem Celular , Núcleo Celular/genética , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-jun/genética , Fatores de Transcrição da Família Snail/biossíntese , Fatores de Transcrição da Família Snail/genética , Proteínas Supressoras de Tumor/genética
18.
Adv Cancer Res ; 126: 11-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25727145

RESUMO

Glycosylation is catalyzed by various glycosyltransferase enzymes which are mostly located in the Golgi apparatus in cells. These enzymes glycosylate various complex carbohydrates such as glycoproteins, glycolipids, and proteoglycans. The enzyme activity of glycosyltransferases and their gene expression are altered in various pathophysiological situations including cancer. Furthermore, the activity of glycosyltransferases is controlled by various factors such as the levels of nucleotide sugars, acceptor substrates, nucleotide sugar transporters, chaperons, and endogenous lectin in cancer cells. The glycosylation results in various functional changes of glycoproteins including cell surface receptors and adhesion molecules such as E-cadherin and integrins. These changes confer the unique characteristic phenotypes associated with cancer cells. Therefore, glycans play key roles in cancer progression and treatment. This review focuses on glycan structures, their biosynthetic glycosyltransferases, and their genes in relation to their biological significance and involvement in cancer, especially cancer biomarkers, epithelial-mesenchymal transition, cancer progression and metastasis, and therapeutics. Major N-glycan branching structures which are directly related to cancer are ß1,6-GlcNAc branching, bisecting GlcNAc, and core fucose. These structures are enzymatic products of glycosyltransferases, GnT-V, GnT-III, and Fut8, respectively. The genes encoding these enzymes are designated as MGAT5 (Mgat5), MGAT3 (Mgat3), and FUT8 (Fut8) in humans (mice in parenthesis), respectively. GnT-V is highly associated with cancer metastasis, whereas GnT-III is associated with cancer suppression. Fut8 is involved in expression of cancer biomarker as well as in the treatment of cancer. In addition to these enzymes, GnT-IV and GnT-IX (GnT-Vb) will be also discussed in relation to cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias/diagnóstico , Neoplasias/terapia , Polissacarídeos/metabolismo , Animais , Progressão da Doença , Glicosilação , Humanos , Metástase Neoplásica , Neoplasias/metabolismo , Polissacarídeos/química
19.
Oncoimmunology ; 2(2): e22945, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23526132

RESUMO

Toll-like receptors (TLRs) have generated an extraordinary amount of interest in cancer research since the last decade. TLRs are a family of pattern recognition receptors that is involved in the host defense against microbial infections. It is well known that the activation of TLRs leads to the production of biological factors that drive inflammatory responses and activate the adaptive immune system. More recently, TLR-mediated signaling pathways have been shown to support tumor cell growth in vitro and in vivo. In this review, we describe recently emerged links between TLR4 and breast cancer oncogenesis, and future perspectives for the targeting of TLR4 in breast cancer therapy.

20.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-594427

RESUMO

Objective To investigate the relationship between NF-?B activation and the expression of metastasis correlation factors.Methods The expression of NF-?B,COX-2,ICAM-1,VCAM-1 and MMP9 in colorectal cancer and normal tissues was detected using Western blotting.Results(1)The expression level of NF-?B was higher in cancer tissues than that in normal tissues(P

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...