Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cancer Immunol Immunother ; 72(2): 351-369, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35895109

RESUMO

BACKGROUND: Immunotherapy is an emerging cancer therapy with potential great success; however, immune checkpoint inhibitor (e.g., anti-PD-1) has response rates of only 10-30% in solid tumor because of the immunosuppressive tumor microenvironment (TME). This affliction can be solved by vascular normalization and TME reprogramming. METHODS: By using the single-cell RNA sequencing (scRNAseq) approach, we tried to find out the reprogramming mechanism that the Fc-VEGF chimeric antibody drug (Fc-VFD) enhances immune cell infiltration in the TME. RESULTS: In this work, we showed that Fc-VEGF121-VEGF165 (Fc-VEGF chimeric antibody drug, Fc-VFD) arrests excess angiogenesis and tumor growth through vascular normalization using in vitro and in vivo studies. The results confirmed that the treatment of Fc-VFD increases immune cell infiltration including cytotoxic T, NK, and M1-macrophages cells. Indeed, Fc-VFD inhibits Lon-induced M2 macrophages polarization that induces angiogenesis. Furthermore, Fc-VFD inhibits the secretion of VEGF-A, IL-6, TGF-ß, or IL-10 from endothelial, cancer cells, and M2 macrophage, which reprograms immunosuppressive TME. Importantly, Fc-VFD enhances the synergistic effect on the combination immunotherapy with anti-PD-L1 in vivo. CONCLUSIONS: In short, Fc-VFD fusion normalizes intratumor vasculature to reprogram the immunosuppressive TME and enhance cancer immunotherapy.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular , Imunoterapia , Antineoplásicos/farmacologia , Imunossupressores/farmacologia
2.
J Clin Lab Anal ; 34(4): e23110, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31733007

RESUMO

BACKGROUND: Despite having chronic gastritis, most people infected by Helicobacter pylori (H. pylori) are asymptomatic and have no specific clinical signs and symptoms. H. pylori infection can be diagnosed by several detection methods. Giemsa stain and rapid urease test (CLO test) are the most performed tests of H. pylori infection at first-line clinical examination because of their simplicity and reliability. However, the sensitivity of CLO test is significantly reduced in patients with atrophic gastritis and intestinal metaplasia, and the weaknesses of Giemsa stain are higher cost and time-consuming. METHODS: The Giemsa stain was modified in several staining solutions and procedures based on the simplified Giemsa technique described by Gray, Wyatt, & Rathbone (1986). The modified Giemsa stain is examined its efficacy and compared with the CLO test using 233 H. pylori-infected patients with gastric disease. RESULTS: The modified Giemsa stain is comparable to the traditional one. Statistical analysis indicated that the modified Giemsa stain obtains greater accuracy in H. pylori-infected patients with gastritis and ulcer than the CLO test (48.1% vs. 43.7%). Moreover, considering the prognosis of different symptoms of gastric diseases, the modified Giemsa stain has a more accurate prognosis than combination symptoms (P = 1.8E-05 vs. P = 5.49E-05). The modified Giemsa stain is confirmed to be better than CLO test using 233 H. pylori-infected patients with gastric disease. CONCLUSIONS: The modified Giemsa stain is more simplified and time-saving than traditional Giemsa stain, which is comparable to the traditional one and is confirmed to be better than CLO test using 233 H. pylori-infected patients with gastric disease. In clinical examination, this modified Giemsa stain can be applied to routine examination and provides quick and accurate diagnosis and prognosis to H. pylori-infected patients with gastric diseases.


Assuntos
Corantes Azur , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/patologia , Urease , Biópsia , Gastrite/microbiologia , Humanos , Úlcera Gástrica/microbiologia , Urease/metabolismo
3.
Reprod Biol Endocrinol ; 16(1): 8, 2018 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-29378615

RESUMO

BACKGROUND: Cystatin C (CST3), a cysteine protease inhibitor in seminal plasma, is expressed in animal uteri. However, its expression in the human female reproductive tract and its effect on human sperm capacitation are unclear. METHODS: The cellular localization of CST3 was observed using immunohistochemistry. The binding of CST3 to sperm was examined using immunocytochemistry. Sperm motility parameters were analyzed using computer-assisted sperm analysis. Sperm capacitation was evaluated by analyzing cholesterol content, protein tyrosine phosphorylation levels, and the acrosome reaction. RESULTS: Immunohistochemical staining demonstrated that CST3 is prominently expressed in the female reproductive tract, including the epithelial lining and cervix and endometrium fluids, particularly at times near ovulation. It can bind to human sperm on the post-acrosomal head region and the mid and principal piece of the tail. CST3 enhances sperm motility and inhibits the signal initiating sperm capacitation, i.e., efflux of cholesterol from the sperm plasma membrane and a late sperm capacitation event, i.e., the increase in the sperm protein tyrosine phosphorylation. The suppressive trend on sperm acrosome reaction further supports CST3's ability to inhibit sperm capacitation. CONCLUSIONS: These findings suggest that cervical CST3 may prevent precocious capacitation and acrosome reaction, thus preserving sperm fertilizing ability before it reaches the fallopian tube. Additionally, CST3 may help sperm enter the upper reproductive tract by enhancing sperm motility.


Assuntos
Cistatina C/fisiologia , Capacitação Espermática/fisiologia , Reação Acrossômica , Colo do Útero/metabolismo , Cistatina C/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Fosforilação , Interações Espermatozoide-Óvulo , Útero/metabolismo
4.
Environ Toxicol ; 32(11): 2379-2391, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28722353

RESUMO

Burning incense to worship deities is a popular religious ritual in large parts of Asia, and is a popular custom affecting more than 1.5 billion adherents. Due to incomplete combustion, burning incense has been well recognized to generate airborne hazards to human health. However, the correlation between burning incense and lung cancer in epidemiological studies remains controversy. Therefore, we speculated that some unknown materials in incense smoke are involved in the initiation or progression of lung cancer. Based on this hypothesis, we identified a major compound auramine O (AuO) from the water-soluble fraction of incense burned condensate using mass spectrometry. AuO is commonly used in incense manufacture as a colorant. Due to thermostable, AuO released from burned incenses becomes an unexpected air pollutant. AuO is classified as a Group 2B chemical by the International Agency of Research on Cancer (IARC), however, the damage of AuO to the respiratory system remains elusive. Our study revealed that AuO has no apparent effect on malignant transformation; but, it dramatically promotes lung cancer malignancy. AuO accumulates in the nucleus and induces the autophagy activity in lung tumor cells. AuO significantly enhances migration and invasive abilities and the in vitro and in vivo stemness features of lung tumor cells through activating the expression of aldehyde dehydrogenase family 1 member A1 (ALDH1A1), and ALDH1A1 knockdown attenuates AuO-induced autophagy activity and blocks AuO-induced lung tumor malignancy. In conclusion, we found that AuO, an ingredient of incense smoke, significantly increases the metastatic abilities and stemness characters of lung tumor cells through the activation of ALDH1A1, which is known to be associated with poor outcome and progression of lung cancer. For public health, reducing or avoiding the use of AuO in incense is recommended.


Assuntos
Adenocarcinoma/patologia , Poluentes Atmosféricos/toxicidade , Benzofenoneídio/toxicidade , Corantes/toxicidade , Neoplasias Pulmonares/patologia , Fumaça/efeitos adversos , Adenocarcinoma/induzido quimicamente , Adenocarcinoma de Pulmão , Poluentes Atmosféricos/análise , Poluição do Ar em Ambientes Fechados , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Linhagem Celular Tumoral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Humanos , Neoplasias Pulmonares/induzido quimicamente , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/patologia , Retinal Desidrogenase , Fumaça/análise , Esferoides Celulares/patologia
5.
Mol Carcinog ; 55(5): 499-513, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-25728212

RESUMO

Growth differentiation factor-10 (GDF10), commonly referred as BMP3b, is a member of the transforming growth factor-ß (TGF-ß) superfamily. GDF10/BMP3b has been considered as a tumor suppressor, however, little is known about the molecular mechanism of its roles in tumor suppression in oral cancer. Clinical significance of GDF10 downregulation in oral squamous cell carcinoma (OSCC) was evaluated using three independent cohorts of OSCC patients. The molecular mechanisms of GDF10 in the suppression of cell survival, cell migration/invasion and epithelial-mesenchymal transition (EMT) were investigated by using oral cancer cell lines. The present study shows that GDF10 is downregulated during oral carcinogenesis, and GDF10 expression is also an independent risk factor for overall survival of OSCC patients. Overexpression of GDF10 attenuates cell proliferation, transformation, migration/invasion, and EMT. GDF10-inhibited EMT is mediated by ERK signaling but not by typical TGF-ß signaling. In addition, overexpression of GDF10 promotes DNA damage-induced apoptosis and sensitizes the response to all-trans retinoic acid (ATRA) and camptothecin (CPT). Intriguingly, the expression of GDF10 is induced by type III TGF-ß receptor (TGFBR3) through TGF-ß-SMAD2/3 signaling. Our findings suggest that TGFBR3 is an upstream activator of GDF10 expression and they share the same signaling to inhibit EMT and migration/invasion. These results support that GDF10 acts as a hinge to collaborate with TGFBR3 in the transition of EMT-MET program. Taken together, we illustrated the clinical significance and the molecular mechanisms of tumor-suppressive GDF10 in OSCC.


Assuntos
Carcinoma de Células Escamosas/patologia , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Fator 10 de Diferenciação de Crescimento/metabolismo , Neoplasias Bucais/patologia , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Fator 10 de Diferenciação de Crescimento/genética , Humanos , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Prognóstico , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Análise de Sobrevida
6.
Clin Lab ; 61(11): 1635-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26731987

RESUMO

BACKGROUND: Targeted therapy is typically used to treat colorectal cancer (CRC). The epidermal growth factor receptor (EGFR) was recognized as a potential therapeutic target. Does the EGFR protein express consistently using different monoclonal antibodies in clinics? METHODS: One hundred and sixty-four patients (mean age 61.80 ± 12.78 years) who suffered from CRC were selected at Mackay Memorial Hospital in Taiwan. Formalin-fixed and paraffin-embedded tissue sections from all patients were tested simultaneously using two commercial antibodies, Dako-EGFR (mouse monoclonal anti-EGFR clone 2-18C9, pharmDx) and NCL-EGFR (NCL-EGFR-384, Novocastra) monoclonal antibodies, to study the commutability or equality of the qualities of EGFR expression by standard immunohistochemistry (IHC) procedures. RESULTS: The EGFR expressions that were obtained by IHC staining using different monoclonal antibodies with Dako-EGFR (46.95%) and NCL-EGFR (32.32%) were fairly concordant. CONCLUSIONS: Although IHC is a convenient and feasible method for detecting the expression of EGFR, it yields controversial staining results concerning EGFR expression using various commercial antibodies in a CRC tumor section.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias Colorretais/metabolismo , Receptores ErbB/metabolismo , Idoso , Neoplasias Colorretais/imunologia , Receptores ErbB/imunologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Taiwan
7.
Int J Cancer ; 135(4): 809-19, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24477565

RESUMO

SOX2 is a transcription factor essential for self-renewal and pluripotency of embryonic stem cells. Recently, SOX2 was found overexpressed in the majority of the lung squamous cell carcinoma (SQC), in which it acts as a lineage-survival oncogene. However, downstream targets/pathways of SOX2 in lung SQC cells remain to be identified. Here, we show that BMP4 is a downstream target of SOX2 in lung SQC. We found that SOX2-silencing-mediated inhibition of cell growth was accompanied by upregulation of BMP4 mRNA and its protein expression. Meta-analysis with 293 samples and qRT-PCR validation with 73 clinical samples revealed an inversely correlated relationship between levels of SOX2 and BMP4 mRNA, and significantly lower mRNA levels in tumor than in adjacent normal tissues. This was corroborated by immunohistochemistry analysis of 35 lung SQC samples showing lower BMP4 protein expression in tumor tissues. Cell-based experiments including siRNA transfection, growth assay and flow cytometry assay, further combined with a xenograft tumor model in mice, revealed that reactivation of BMP4 signaling could partially account for growth inhibition and cell cycle arrest in lung SQC cells upon silencing SOX2. Finally, chromatin immunoprecipitation analysis and luciferase reporter assay revealed that SOX2 could negatively regulate BMP4 promoter activity, possibly through binding to the promoter located in the first intron region of BMP4. Collectively, our findings suggest that BMP4 could act as a tumor suppressor and its downregulation by elevated SOX2 resulting in enhanced growth of lung SQC cells.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Pulmonares/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Escamosas/genética , Bases de Dados Genéticas , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/genética , Camundongos , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Transdução de Sinais
8.
Med Mycol ; 52(3): 276-84, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24625676

RESUMO

Favus is a distinctive form of infection that is caused by exclusively dermatophytes. Its clinical presentation is characterized by scutula, which are concave, thick fungal crusts. The best-known examples of human scalp favus are caused by Trichophyton schoenleinii and those of mouse favus are caused by T. quinckeanum. However, other dermatophytes, such as T. violaceum, T. verrucosum, Microsporum audouinii, M. gallinae, M. gypseum, and M. canis, have been reported sporadically to cause favic lesions. Favus on cats has rarely been mentioned in the literature, and the pathogens with which it has been associated are, for the most part, unknown. Here, we examine four cat favus cases, focusing on clinical presentations and histopathological features. In all cases the etiologic agent was identified as M. incurvatum based on its morphological characteristics and sequences of internal transcribed spacers (ITS) of nuclear ribosomal DNA. Phylogenetic analysis using the neighbor-joining method, which is based on ITS, showed that these four isolates belonged to two strains of M. incurvatum; one strain was a new combination from the basionym Nannizzia incurvata.


Assuntos
Doenças do Gato/diagnóstico , Doenças do Gato/patologia , Microsporum/classificação , Microsporum/isolamento & purificação , Tinha Favosa/veterinária , Animais , Doenças do Gato/microbiologia , Gatos , Análise por Conglomerados , DNA Fúngico/química , DNA Fúngico/genética , DNA Espaçador Ribossômico/química , DNA Espaçador Ribossômico/genética , Feminino , Histocitoquímica , Masculino , Camundongos , Microscopia , Microsporum/citologia , Microsporum/genética , Dados de Sequência Molecular , Filogenia , Análise de Sequência de DNA , Tinha Favosa/diagnóstico , Tinha Favosa/microbiologia , Tinha Favosa/patologia
9.
Tumour Biol ; 34(5): 2557-64, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23625657

RESUMO

hPuf-A is a member of RNA-binding PUF family that regulates mRNA translation. Redistribution of hPuf-A from the nucleolus to the nucleoplasm upon genotoxic stress modulates the poly(ADP-ribosyl)ation activity of PARP-1. Here, we report a novel function of hPuf-A involved in promoting breast cancer progression. Immunohistochemical studies showed higher expression levels of hPuf-A in stage I, II, III, and IV breast cancer specimens in contrast with those of hPuf-A in ductal carcinoma in situ. The presence of hPuf-A is highly associated with colony formation capacities in breast cancer T47D and MDA-MB-231 cells. Xenograft growth of hPuf-A-silenced and hPuf-A overexpressing MDA-MB-231 cells in nude mice was substantially in concert with colony formation capacities. This promoting effect of hPuf-A in tumorigenesis might be correlated with the regulation of its associated mRNAs, such as RbAp48 and DDX3. Collectively, hPuf-A may have diagnostic values in breast cancer progression.


Assuntos
Neoplasias da Mama/metabolismo , Carcinogênese/metabolismo , Carcinoma Intraductal não Infiltrante/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Neoplasias da Mama/patologia , Carcinogênese/patologia , Carcinoma Intraductal não Infiltrante/secundário , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Antígenos de Histocompatibilidade Menor , Estadiamento de Neoplasias , Transplante de Neoplasias , Proteínas de Ligação a RNA/metabolismo , Carga Tumoral , Regulação para Cima
10.
Cell Death Dis ; 14(3): 199, 2023 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-36927870

RESUMO

During hypoxia, FUNDC1 acts as a mitophagy receptor and accumulates at the ER (endoplasmic reticulum)-mitochondria contact sites (EMC), also called mitochondria-associated membranes (MAM). In mitophagy, the ULK1 complex phosphorylates FUNDC1(S17) at the EMC site. However, how mitochondria sense the stress and send the signal from the inside to the outside of mitochondria to trigger mitophagy is still unclear. Mitochondrial Lon was reported to be localized at the EMC under stress although the function remained unknown. In this study, we explored the mechanism of how mitochondrial sensors of hypoxia trigger and stabilize the FUNDC1-ULK1 complex by Lon in the EMC for cell survival and cancer progression. We demonstrated that Lon is accumulated in the EMC and associated with FUNDC1-ULK1 complex to induce mitophagy via chaperone activity under hypoxia. Intriguingly, we found that Lon-induced mitophagy is through binding with mitochondrial Na+/Ca2+ exchanger (NCLX) to promote FUNDC1-ULK1-mediated mitophagy at the EMC site in vitro and in vivo. Accordingly, our findings highlight a novel mechanism responsible for mitophagy initiation under hypoxia by chaperone Lon in mitochondria through the interaction with FUNDC1-ULK1 complex at the EMC site. These findings provide a direct correlation between Lon and mitophagy on cell survival and cancer progression.


Assuntos
Proteínas de Membrana , Mitofagia , Humanos , Fosforilação , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Hipóxia/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo
11.
JCI Insight ; 7(23)2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36264639

RESUMO

Docetaxel (DTX) combined with cisplatin and 5-fluorouracil has been used as induction chemotherapy for head and neck squamous cell carcinoma (HNSCC). However, the development of acquired resistance remains a major obstacle to treatment response. Tumor-associated macrophages are associated with chemotherapeutic resistance. In the present study, increased infiltration of macrophages into the tumor microenvironment (TME) was significantly associated with shorter overall survival and increased resistance to chemotherapeutic drugs, particularly DTX, in patients with HNSCC. Macrophage coculture induced expression of intercellular adhesion molecule 1 (ICAM1), which promotes stemness and the formation of polyploid giant cancer cells, thereby reducing the efficacy of DTX. Both genetic silencing and pharmacological inhibition of ICAM1 sensitized HNSCC to DTX. Macrophage secretion of IL-1ß was found to induce tumor expression of ICAM1. IL-1ß neutralization and IL-1 receptor blockade reversed DTX resistance induced by macrophage coculture. IL-1ß activated superoxide dismutase 2 and inhibited catalase, thereby modulating intracellular levels of ROS and inducing ICAM1 expression. Arsenic trioxide (ATO) reduced macrophage infiltration into the TME and impaired IL-1ß secretion by macrophages. The combinatorial use of ATO enhanced the in vivo efficacy of DTX in a mouse model, which may provide a revolutionary approach to overcoming acquired therapeutic resistance in HNSCC.


Assuntos
Docetaxel , Neoplasias de Cabeça e Pescoço , Molécula 1 de Adesão Intercelular , Interleucina-1beta , Carcinoma de Células Escamosas de Cabeça e Pescoço , Animais , Camundongos , Docetaxel/farmacologia , Docetaxel/uso terapêutico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Molécula 1 de Adesão Intercelular/genética , Macrófagos , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Microambiente Tumoral , Humanos , Interleucina-1beta/metabolismo , Transdução de Sinais
12.
Reprod Biol Endocrinol ; 9: 38, 2011 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-21426587

RESUMO

BACKGROUND: SERPINE2, also known as protease nexin-1, belongs to the serine protease inhibitor (SERPIN) superfamily. It is one of the potent SERPINs that modulates the activity of plasminogen activators (PAs). PAs and their SERPIN inhibitors, such as SERPINB2 and SERPINE1, were expressed in the human endometrium and were implicated in implantation. However, expression data about SERPINE2 in the human endometrium is still unknown. Thus, we conducted an investigation to reveal the spatiotemporal and cellular expression of SERPINE2 in the human uterus during the menstrual cycle. METHODS: Seven patients who underwent a hysterectomy and samples of 120 archived patients' endometrial curettage or parts of the uterus that were formalin-fixed and embedded in paraffin. Western blotting was performed to evaluate the specificity and sensitivity of the antibody. Immunohistochemistry was conducted to localize the SERPINE2 expression site. Quantitative analysis was conducted to evaluate expression levels of SERPINE2 in various sub-phases of the menstrual cycle. RESULTS: The SERPINE2 protein was primarily detected in the uterine fluid during the mid- and late-secretory phases of the menstrual cycle. It was predominantly expressed in the luminal and glandular epithelium, less in the myometrium, and only dispersedly in certain stromal cells throughout the menstrual cycle. A quantitative analysis of expression levels of SERPINE2 in the glandular epithelium revealed that it was highly expressed in the endometrium during the secretory phase compared to the proliferative phase. CONCLUSIONS: The SERPINE2 protein is highly expressed in the endometrium during the secretory phase, indicating that it may participate in tissue remodeling involved in implantation.


Assuntos
Fase Luteal/metabolismo , Serpina E2/biossíntese , Líquidos Corporais/química , Endométrio/metabolismo , Feminino , Humanos , Útero/química
13.
Redox Biol ; 34: 101571, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32446175

RESUMO

Recurrence and metastasis remain the major cause of cancer mortality. Even for early-stage lung cancer, adjuvant chemotherapy yields merely slight increase to patient survival. EF-hand domain-containing protein D2 (EFHD2) has recently been implicated in recurrence of patients with stage I lung adenocarcinoma. In this study, we investigated the correlation between EFHD2 and chemoresistance in non-small cell lung cancer (NSCLC). High expression of EFHD2 was significantly associated with poor overall survival of NSCLC patients with chemotherapy in in silica analysis. Ectopic EFHD2 overexpression increased cisplatin resistance, whereas EFHD2 knockdown improved chemoresponse. Mechanistically, EFHD2 induced the production of NADPH oxidase 4 (NOX4) and in turn the increase of intracellular reactive oxygen species (ROS), consequently activating membrane expression of the ATP-binding cassette subfamily C member 1 (ABCC1) for drug efflux. Non-steroidal anti-inflammatory drug (NSAID) ibuprofen suppressed EFHD2 expression by leading to the proteasomal and lysosomal degradation of EFHD2 through a cyclooxygenase (COX)-independent mechanism. Combining ibuprofen with cisplatin enhanced antitumor responsiveness in a murine xenograft model in comparison with the individual treatment. In conclusion, we demonstrate that EFHD2 promotes chemoresistance through the NOX4-ROS-ABCC1 axis and therefore developing EFHD2-targeting strategies may offer a new avenue to improve adjuvant chemotherapy of lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Proteínas de Ligação ao Cálcio , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Camundongos , Proteínas Associadas à Resistência a Múltiplos Medicamentos , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
14.
Cancer Lett ; 474: 138-150, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31987921

RESUMO

Mitochondrial Lon is a chaperone protein whose upregulation increases the production of mitochondrial reactive oxygen species (ROS). However, there is a lack of information in detail on how mitochondrial Lon regulates cancer metastasis through ROS production in the tumor microenvironment (TME). Our results show that elevated Lon promotes epithelial-mesenchymal transition (EMT) via ROS-dependent p38 and NF-κB-signaling. We further identified pyrroline-5-carboxylate reductase 1 (PYCR1) as a client of chaperone Lon, which induces mitochondrial ROS and EMT by Lon. Mitochondrial Lon induces ROS-dependent production of inflammatory cytokines, such as TGF-ß, IL-6, IL-13, and VEGF-A, which consequently activates EMT, angiogenesis, and M2 macrophage polarization. In addition, Lon expression is induced upon the activation and M2 polarization of macrophages, which further promotes M2 macrophages to enhance the immunosuppressive microenvironment and metastatic behaviors in the TME. This raises the possibility that manipulation of the mitochondrial redox balance in the TME may serve as a therapeutic strategy to improve T cell function in cancer immunotherapy.


Assuntos
Proteases Dependentes de ATP/metabolismo , Neoplasias Pulmonares/secundário , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Neoplasias Bucais/patologia , Estresse Oxidativo , Pirrolina Carboxilato Redutases/metabolismo , Proteases Dependentes de ATP/genética , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proliferação de Células , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Ativação de Macrófagos/imunologia , Masculino , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Neoplasias Bucais/imunologia , Neoplasias Bucais/metabolismo , Prognóstico , Pirrolina Carboxilato Redutases/genética , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , delta-1-Pirrolina-5-Carboxilato Redutase
15.
Audiol Neurootol ; 13(4): 239-46, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18259076

RESUMO

INTRODUCTION: Obliteration of the mastoid cavities with auricular cartilage is a frequently used method to minimize the open cavity problem in cholesteatoma surgery. However, the method of cartilage preparation and histopathologic changes of the grafted cartilage in patients receiving mastoid obliteration are rarely reported. Hence, the authors developed rabbit tympanic bulla obliteration with auricular cartilage as an animal model and studied the effects of perichondrium preservation on the grafted cartilage. MATERIALS AND METHODS: Auricular cartilage with or without perichondrium was prepared and cut into small pieces to obliterate rabbit tympanic bullae. Four weeks after surgery, the viable chondrocyte ratio indicated by the number of viable chondrocytes divided by the total number of chondrocytes, the microvascular density shown by CD31-labeled vessels, and the chondrogenesis ratio represented by the ratio of the cross-sectional areas of the newly formed cartilage and the originally grafted cartilage were calculated and compared. RESULTS: The viable chondrocyte ratio was 49.21 +/- 10.17% in the perichondrium-preserved group (n = 12) and 35.46 +/- 3.96% in the perichondrium-removed group (n = 12, p = 0.001). The CD31 microvascular density was significantly higher in the perichondrium-preserved group than in the perichondrium-removed group (167.77 +/- 15.83 vs. 77.17 +/- 19.67 microvessels/mm(2), p < 0.001). The chondrogenesis ratios were 27.58 +/- 12.44% in the perichondrium-preserved group and 0.45 +/- 0.63% in the perichondrium-removed group (p < 0.001). CONCLUSION: Obliteration of tympanic bullae with perichondrium-preserved cartilage results in faster restoration of circulation, higher survival of chondrocytes and more cartilage regeneration than with perichondrium-removed cartilage.


Assuntos
Cartilagem/transplante , Sobrevivência Celular/fisiologia , Condrócitos/fisiologia , Condrogênese/fisiologia , Orelha Média/cirurgia , Preservação de Tecido/métodos , Animais , Cartilagem/irrigação sanguínea , Contagem de Células , Condrócitos/patologia , Tecido Conjuntivo/patologia , Tecido Conjuntivo/fisiologia , Orelha Média/irrigação sanguínea , Orelha Média/patologia , Feminino , Técnicas Imunoenzimáticas , Masculino , Microcirculação/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Coelhos , Regeneração/fisiologia
16.
Sci Rep ; 8(1): 1456, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29348465

RESUMO

A correction to this article has been published and is linked from the HTML version of this paper. The error has been fixed in the paper.

17.
Cell Death Dis ; 9(6): 697, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29899330

RESUMO

Mitochondrial Lon is a multi-function matrix protease with chaperone activity. However, little literature has been undertaken into detailed investigations on how Lon regulates apoptosis through its chaperone activity. Accumulating evidences indicate that various stresses induce transportation of p53 to mitochondria and activate apoptosis in a transcription-independent manner. Here we found that increased Lon interacts with p53 in mitochondrial matrix and restrains the apoptosis induced by p53 under oxidative stress by rescuing the loss of mitochondrial membrane potential (Δψm) and the release of cytochrome C and SMAC/Diablo. Increased chaperone Lon hampers the transcription-dependent apoptotic function of p53 by reducing the mRNA expression of p53 target genes. The ATPase mutant (K529R) of chaperone Lon decreases the interaction with p53 and fails to inhibit apoptosis. Furthermore, the chaperone activity of Lon is important for mitochondrial p53 accumulation in an mtHsp70-dependent manner, which is also important to prevent the cytosolic distribution of p53 from proteasome-dependent degradation. These results indicate that the chaperone activity of Lon is important to bind with mitochondrial p53 by which increased Lon suppresses the apoptotic function of p53 under oxidative stress. Furthermore, mitochondrial Lon-mtHsp70 increases the stability/level of p53 through trafficking and retaining p53 in mitochondrial matrix and preventing the pool of cytosolic p53 from proteasome-dependent degradation in vitro and in clinic.


Assuntos
Apoptose , Mitocôndrias/metabolismo , Chaperonas Moleculares/metabolismo , Estresse Oxidativo , Protease La/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Citosol/metabolismo , Humanos , Simulação de Acoplamento Molecular , Neoplasias Bucais/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Estabilidade Proteica , Proteólise , Transcrição Gênica
18.
Oncotarget ; 8(9): 14666-14679, 2017 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-28108734

RESUMO

We studied the potential mechanisms of valproic acid (VPA) in the treatment of glioblastoma multiforme (GBM). Using the human U87, GBM8401, and DBTRG-05MG GBM-derived cell lines, VPA at concentrations of 5 to 20 mM induced G2/M cell cycle arrest and increased the production of reactive oxygen species (ROS). Stress-related molecules such as paraoxonase 2 (PON2), cyclin B1, cdc2, and Bcl-xL were downregulated, but p27, p21 and Bim were upregulated by VPA treatment. VPA response element on the PON2 promoter was localized at position -400/-1. PON2 protein expression was increased in GBM cells compared with normal brain tissue and there was a negative correlation between the expression of PON2 and Bim. These findings were confirmed by the public Bredel GBM microarray (Gene Expression Omnibus accession: GSE2223) and the Cancer Genome Atlas GBM microarray datasets. Overexpression of PON2 in GBM cells significantly decreased intracellular ROS levels, and PON2 expression was decreased after VPA stimulation compared with controls. Bim expression was significantly induced by VPA in GBM cells with PON2 silencing. These observations were further shown in the subcutaneous GBM8401 cell xenograft of BALB/c nude mice. Our results suggest that VPA reduces PON2 expression in GBM cells, which in turn increases ROS production and induces Bim production that inhibits cancer progression via the PON2-Bim cascade.


Assuntos
Arildialquilfosfatase/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Ácido Valproico/farmacologia , Animais , Arildialquilfosfatase/genética , Proteína 11 Semelhante a Bcl-2/genética , Proteína 11 Semelhante a Bcl-2/metabolismo , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , GABAérgicos/farmacologia , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Regiões Promotoras Genéticas/genética , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Sci Rep ; 7(1): 14617, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29097801

RESUMO

Surgery is the only curative treatment for early-stage non-small cell lung cancer (NSCLC) patients. However, approximately one-third of these patients develop recurrence, which remains the main cause of mortality in the postsurgical treatment of NSCLC. Many molecular markers have been proposed to predict recurrence of early-stage disease, but no marker has demonstrated sufficient reliability for clinical application. In the present study, the novel protein EF-hand domain-containing protein D2 (EFHD2) was identified as expressed in highly metastatic tumor cells. EFHD2 increased the formation of protrusive invadopodia structures and cell migration and invasion abilities and promoted the epithelial-to-mesenchymal transition (EMT) character of lung adenocarcinoma cells. We demonstrated that the mechanism of EFHD2 in enhancing EMT occurs partly through inhibition of caveolin-1 (CAV1) for cancer progression. The expression of EFHD2 was significantly correlated with postsurgical recurrence of patients with stage I lung adenocarcinoma in the Kaplan-Meier-plotter cancer database search and our retrospective cohort study (HR, 6.14; 95% CI, 2.40-15.74; P < 0.001). Multivariate Cox regression analysis revealed that EFHD2 expression was an independent clinical predictor for this disease. We conclude that EFHD2 expression is associated with increased metastasis and EMT and could serve as an independent marker to predict postsurgical recurrence of patients with stage I lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Pulmonares/metabolismo , Recidiva Local de Neoplasia/metabolismo , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/cirurgia , Idoso , Biomarcadores Tumorais/metabolismo , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Masculino , Invasividade Neoplásica/fisiopatologia , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos , Análise de Sobrevida
20.
Sci Rep ; 7(1): 17024, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29209046

RESUMO

Cdc7-Dbf4 kinase plays a key role in the initiation of DNA replication and contributes to the replication stress in cancer. The activity of human Cdc7-Dbf4 kinase remains active and acts as an effector of checkpoint under replication stress. However, the downstream targets of Cdc7-Dbf4 contributed to checkpoint regulation and replication stress-support function in cancer are not fully identified. In this work, we showed that aberrant Cdc7-Dbf4 induces DNA lesions that activate ATM/ATR-mediated checkpoint and homologous recombination (HR) DNA repair. Using a phosphoproteome approach, we identified HSP90-S164 as a target of Cdc7-Dbf4 in vitro and in vivo. The phosphorylation of HSP90-S164 by Cdc7-Dbf4 is required for the stability of HSP90-HCLK2-MRN complex and the function of ATM/ATR signaling cascade and HR DNA repair. In clinically, the phosphorylation of HSP90-S164 indeed is increased in oral cancer patients. Our results indicate that aberrant Cdc7-Dbf4 enhances replication stress tolerance by rewiring ATR/ATM mediated HR repair through HSP90-S164 phosphorylation and by promoting recovery from replication stress. We provide a new solution to a subtyping of cancer patients with dominant ATR/HSP90 expression by combining inhibitors of ATR-Chk1, HSP90, or Cdc7 in cancer combination therapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Replicação do DNA , Regulação Neoplásica da Expressão Gênica , Neoplasias Bucais/patologia , Estresse Fisiológico , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Estudos de Casos e Controles , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Seguimentos , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa