Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Biotechnol Lett ; 43(10): 1945-1954, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34448096

RESUMO

Oncolytic therapy is a treatment method used to directly combat tumor cells by modifying the genes of naturally occurring low pathogenic viruses to form "rhizobia" virus. By taking the advantage of abnormal signal pathways in cancer cells, it selectively replicates in tumor cells leading to tumor cell lysis and death. At present, clinical studies widely employ biomolecular technology to transform oncolytic viruses to exert stronger oncolytic effects and reduce their adverse reactions. This review summarizes the current progresses and the molecular mechanism of oncolytic viruses towards tumor treatment and management.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Humanos , Camundongos
2.
Gen Physiol Biophys ; 39(3): 277-283, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32525821

RESUMO

With the increasing incidence of premature ovarian failure (POF) seriously threaten the women's health. Whether cryptotanshinone decreased the granulosa cell apoptosis to improve the POF would be explored. POF mice were conducted with intraperitoneal injection of cyclophosphamide and then treated with cryptotanshinone. The body weight and ovarian weight were recorded. The estrus was detected by vaginal smears. The pathological changes of ovarian were observed with hematoxylin and eosin staining. ELISA assay analyzed the levels of LH, FSH, AMH, E2 and AzpAB in mice serum. The expression of Bcl-2, Bax, KI67 and PCNA in ovarian tissues was detected by Western blot analysis and KI67 expression was also determined by immunohistochemistry. The body weight and ovarian weight were decreased and the pathological results of ovarian were worsen in POF mice. The estrus was decreased in POF mice. The levels of LH, FSH and AzpAB were increased and the levels of AMH and E2 were decreased in POF mice serum. The expression of Bcl-2, KI67 and PCNA was decreased and Bax expression was increased in ovarian tissues of POF mice. Those changes affected by cyclophosphamide could be reversed by cryptotanshinone. Cryptotanshinone could decrease the granulosa cell apoptosis to restore ovarian function.


Assuntos
Apoptose/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Fenantrenos/uso terapêutico , Insuficiência Ovariana Primária/tratamento farmacológico , Animais , Feminino , Células da Granulosa/citologia , Camundongos , Ovário/efeitos dos fármacos
3.
Mol Cancer ; 18(1): 156, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31694640

RESUMO

BACKGROUND: Aspartate ß-hydroxylase (ASPH) is silent in normal adult tissues only to re-emerge during oncogenesis where its function is required for generation and maintenance of malignant phenotypes. Exosomes enable prooncogenic secretome delivering and trafficking for long-distance cell-to-cell communication. This study aims to explore molecular mechanisms underlying how ASPH network regulates designated exosomes to program development and progression of breast cancer. METHODS: Stable cell lines overexpressing or knocking-out of ASPH were established using lentivirus transfection or CRISPR-CAS9 systems. Western blot, MTT, immunofluorescence, luciferase reporter, co-immunoprecipitation, 2D/3-D invasion, tube formation, mammosphere formation, immunohistochemistry and newly developed in vitro metastasis were applied. RESULTS: Through physical interactions with Notch receptors, ligands (JAGs) and regulators (ADAM10/17), ASPH activates Notch cascade to provide raw materials (especially MMPs/ADAMs) for synthesis/release of pro-metastatic exosomes. Exosomes orchestrate EMT, 2-D/3-D invasion, stemness, angiogenesis, and premetastatic niche formation. Small molecule inhibitors (SMIs) of ASPH's ß-hydroxylase specifically/efficiently abrogated in vitro metastasis, which mimics basement membrane invasion at primary site, intravasation/extravasation (transendothelial migration), and colonization/outgrowth at distant sites. Multiple organ-metastases in orthotopic and tail vein injection murine models were substantially blocked by a specific SMI. ASPH is silenced in normal adult breast, upregulated from in situ malignancies to highly expressed in invasive/advanced ductal carcinoma. Moderate-high expression of ASPH confers more aggressive molecular subtypes (TNBC or Her2 amplified), early recurrence/progression and devastating outcome (reduced overall/disease-free survival) of breast cancer. Expression profiling of Notch signaling components positively correlates with ASPH expression in breast cancer patients, confirming that ASPH-Notch axis acts functionally in breast tumorigenesis. CONCLUSIONS: ASPH-Notch axis guides particularly selective exosomes to potentiate multifaceted metastasis. ASPH's pro-oncogenic/pro-metastatic properties are essential for breast cancer development/progression, revealing a potential target for therapy.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Exossomos/metabolismo , Proteínas de Membrana/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Musculares/metabolismo , Proteoma , Receptores Notch/metabolismo , Animais , Biomarcadores , Comunicação Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Feminino , Genes Reporter , Xenoenxertos , Humanos , Imuno-Histoquímica , Ligantes , Metaloproteinases da Matriz/metabolismo , Camundongos , Modelos Biológicos , Metástase Neoplásica , Fenótipo , Transdução de Sinais
4.
Carcinogenesis ; 38(7): 748-755, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28419191

RESUMO

Malignant cancer cell uncontrolled growth depends on the persistent nutrient availability such as glucose and amino acids, which is required for cancer cell energetic and biosynthetic pathways. As a nuclear hormone receptor, peroxisome-proliferator-activated receptor δ (PPARδ) plays a critical role in inflammation and cancer, however, it is still unclear the regulatory mechanism of PPARδ on cancer cell metabolism. Here, we found that PPARδ directly regulated neutral amino acid transporter SLC1-A5 (solute carrier family 1 member 5) and glucose transporter-1 (Glut1) gene transcription, leading to uptake of glucose and amino acid, activation of mTOR signaling, and tumor progression. In contrast, silence of PPARδ or its antagonist inhibited this event. More importantly, PPARδ promoted cancer cell metabolic reprogramming resulting in chemoresistance, which was alleviated by PPARδ antagonist. These findings revealed a novel mechanism of PPARδ-mediated tumor progression, which provided a potential strategy for cancer treatment.


Assuntos
Sistema ASC de Transporte de Aminoácidos/genética , Transportador de Glucose Tipo 1/genética , Antígenos de Histocompatibilidade Menor/genética , Neoplasias/metabolismo , PPAR gama/genética , Serina-Treonina Quinases TOR/genética , Sistema ASC de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Animais , Vias Biossintéticas , Metabolismo Energético/genética , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Células HCT116 , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Antígenos de Histocompatibilidade Menor/metabolismo , Neoplasias/genética , Neoplasias/patologia , PPAR gama/metabolismo , Transcrição Gênica
5.
J Cell Biochem ; 118(6): 1556-1562, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27918085

RESUMO

Abundant nutrient availability including glucose and amino acids plays an important role in maintaining cancer cell energetic and biosynthetic pathways. As a nuclear receptor, peroxisome-proliferator-activated receptor α (PPARα) regulates inflammation and cancer progression, however, it is still unclear the interaction of PPARα with the cancer cell glucose metabolism. Here we found that PPARα reduced Glut1 (Glucose transporter 1) protein and gene levels in HCT-116, SW480, HeLa, and MCF-7 cancer cell lines. In contrast, silenced PPARα reversed this event. Further analysis shows that PPARα directly targeted the consensus PPRE motif of Glut1 promoter region resulting in Glut1 transcription repression. PPARα-mediated Glut1 transcription repression led to decreased influx of glucose in cancer cells. These findings revealed a novel mechanism of PPARα-mediated cancer cell Glut1 transcription repression. J. Cell. Biochem. 118: 1556-1562, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Regulação para Baixo , Transportador de Glucose Tipo 1/genética , Neoplasias/metabolismo , PPAR alfa/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , Neoplasias/genética , Regiões Promotoras Genéticas
6.
Carcinogenesis ; 37(2): 215-222, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718225

RESUMO

Dysregulated expression of epidermal growth factor receptor (EGFR) has been implicated in many cancer events, while peroxisome proliferator-activated receptor γ (PPARγ) negatively regulates cancer progression. The molecular mechanism of EGFR interaction with PPARγ is still unclear. Here, we found that nuclear EGFR induced phosphorylation of PPARγ at Tyr-74 leading to PPARγ ubiquitination and degradation by mouse double minute 2 (MDM2) ubiquitin ligase. PPARγ degradation by EGFR/MDM2 signaling resulted in accumulation of nuclear factor-kappaB (NF-κB)/p65 protein levels and increasing NF-κB activation. In contrast, PPARγ-Y74A mutant reversed this event. Moreover, PPARγ-Y74A mutant suppressed cell proliferation and increased chemotherapeutic agent-induced cancer cell sensitivity. Importantly, the clinical findings show that the nuclear phosphorylation of PPARγ-Y74 and EGFR expression in colonic cancer tissues was higher than that in control normal tissues. Thus, our study revealed a novel molecular mechanism that nuclear EGFR/NF-κB signaling promoted cell proliferation by destructing PPARγ function, which provides a novel strategy for cancer treatment.


Assuntos
Neoplasias do Colo/patologia , Receptores ErbB/metabolismo , NF-kappa B/metabolismo , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Ativação Enzimática/fisiologia , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia
7.
Hum Cell ; 37(4): 931-943, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38814516

RESUMO

Immunosuppressive regulatory cells (IRCs) play important roles in negatively regulating immune response, and are mainly divided into myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). Large numbers of preclinical and clinical studies have shown that inhibition or reduction of IRCs could effectively elevate antitumor immune responses. However, several studies also reported that excessive inhibition of IRCs function is one of the main reasons causing the side effects of cancer immunotherapy. Therefore, the reasonable regulation of IRCs is crucial for improving the safety and efficiency of cancer immunotherapy. In this review, we summarised the recent research advances in the cancer immunotherapy by regulating the proportion of IRCs, and discussed the roles of IRCs in regulating tumour immune evasion and drug resistance to immunotherapies. Furthermore, we also discussed how to balance the potential opportunities and challenges of using IRCs to improve the safety of cancer immunotherapies.


Assuntos
Imunoterapia , Células Supressoras Mieloides , Neoplasias , Linfócitos T Reguladores , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Imunoterapia/métodos , Linfócitos T Reguladores/imunologia , Células Supressoras Mieloides/imunologia , Evasão Tumoral/imunologia , Resistencia a Medicamentos Antineoplásicos/imunologia , Animais , Terapia de Imunossupressão
8.
Onkologie ; 36(10): 598-601, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24107916

RESUMO

The peroxisome proliferator-activated receptor-γ (PPARγ) belongs to the nuclear hormone receptor superfamily, and is expressed in adipose tissue, skeletal muscle, spleen, heart, liver, placenta, lung, and ovary. PPARγ is a critical regulator of inflammation, adipocyte differentiation, glucose homeostasis, and tumorigenesis. The mechanism of PPARγ on tumor suppression is still unclear, but plenty of evidence shows that PPARγ provides new therapeutic targets for cancer. Here we give a review of how PPARγ and its ligands regulate tumorigenesis by different pathways.


Assuntos
Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , PPAR gama/metabolismo , Transdução de Sinais , Animais , Humanos
9.
Behav Brain Res ; 450: 114455, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37148915

RESUMO

BACKGROUND: Cataract has been shown to be associated with an increased risk of cognitive impairment. However, the results of previous studies have been inconsistent. This systematic review and meta-analysis aimed to investigate the association between cataract and the incidence of cognitive impairment in older adults. METHODS: A comprehensive search of electronic databases from inception to January 2023 was performed to identify relevant studies. Data were extracted from eligible studies and a meta-analysis was performed to calculate the pooled hazard ratio (HR) and 95% confidence interval (CI). RESULTS: We included 13 studies with 25 study arms involving a total of 798,694 participants. Compared with participants without cataract, those with cataract had a higher risk of developing all-cause dementia (pooled HR: 1.22; 95 % CI: 1.08-1.38; I2 =86 %; 9 studies), Alzheimer's disease dementia (pooled HR: 1.18; 95 % CI: 1.07-1.30; I2 =0 %; 9 studies), vascular dementia (pooled HR: 1.21; 95 % CI: 1.02-1.43; I2 =77 %;3 studies) and mild cognitive impairment (pooled HR: 1.30; 95% CI: 1.13-1.50; I2 =0%;2 studies). There was no significant association between cataract and mixed dementia (pooled HR: 1.03; 95 % CI: 0.52-2.04; I2 =78 %;2 studies). We assessed the risk of bias of the included studies using the Newcastle-Ottawa Scale and found that most of them had a low or moderate risk of bias. The number of studies in each meta-analysis ranged from two to nine, with more studies available for all-cause dementia and Alzheimer's disease dementia than for vascular dementia and mixed dementia. CONCLUSIONS: The findings suggest that cataract may be associated with cognitive impairment in older adults. However, the causal relationship between cataract and cognition remains unclear and requires further investigation.


Assuntos
Doença de Alzheimer , Catarata , Disfunção Cognitiva , Demência Vascular , Humanos , Idoso , Doença de Alzheimer/complicações , Demência Vascular/complicações , Incidência , Disfunção Cognitiva/epidemiologia , Disfunção Cognitiva/complicações , Catarata/complicações , Catarata/epidemiologia
10.
Curr Mol Med ; 23(10): 1028-1036, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36214301

RESUMO

The CD99 antigen is a transmembrane protein expressed in a broad variety of tissues, particularly in hematopoietic cells, thymus, endothelial cells, etc. It participates in several crucial biological processes, including cell adhesion, migration, death, differentiation, and inflammation. CD99 has shown oncogenic or tumor suppressor roles in different types of cancer. Therefore, it has been used as a biomarker and therapeutic target for several types of cancer. Moreover, it has also been reported to be involved in several critical immune processes, such as T cell activation and differentiation, dendritic cell differentiation, and so on. Hence, CD99 may have potential values in cancer immunotherapy. Anti-CD99 antibodies have shown therapeutic effects on certain types of cancer, especially on Ewing sarcoma and T cell acute lymphoblastic leukemia (ALL). This review summarizes the recent progress of CD99 in cancer research and targeting therapies, especially in cancer immunotherapy, which may help researchers understand the crucial roles of CD99 in cancer development and design new therapeutic strategies.


Assuntos
Antígeno 12E7 , Imunoterapia , Neoplasias , Adesão Celular , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Neoplasias/terapia , Humanos
11.
Open Med (Wars) ; 17(1): 1084-1091, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35799600

RESUMO

The tumor microenvironment is highly immunosuppressive. The genetically modified oncolytic vaccinia virus (OVV) is a promising vector for cancer immunotherapy. The aim of the present study was to assess the antitumor effects of human interleukin-2 (hIL2)-armed OVV in vitro. The hIL2 gene was inserted into a thymidine kinase and the viral growth factor double deleted oncolytic VV (VVDD) to generate recombinant hIL2-armed OVV (rVVDD-hIL2). Viral replication capacity in A549 cells was quantified by plaque titration on CV-1 cells. Production of hIL2 in cancer cells infected by rVVDD-hIL2 was measured by enzyme-linked immunosorbent assay. Finally, 3-(4,5-dimethylthiazol-2-yl)-5-(3-arboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay was performed to assess the antitumor effects of rVVDD-hIL2. The results showed that rVVDD-hIL2 viral particles expressed increasing levels of hIL2 in human and murine cancer cell lines with growing multiplicities of infection (MOIs). The insertion of the hIL2 gene did not impair the replication capacity of VV, and the rVVDD-hIL2 virus killed cancer cells efficaciously. The lytic effects of the recombinant oncolytic virus on tumor cells increased with the growing MOIs. In conclusion, these findings suggest that hIL2-armed VVDD effectively infects and lyses tumor cells, with high expression of hIL2.

12.
Hum Cell ; 35(2): 441-447, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35032297

RESUMO

Engineered T cells expressing chimeric antigen receptors (CARs) with tumor specificity have shown remarkable therapeutic effects on hematologic malignancies. However, CAR-T cells are less effective on solid tumors mainly due to the weak persistence of CAR-T cells, which might be caused by T cell death. Significant activation-induced cell death (AICD) of CAR-T cells was triggered by repeated antigen stimulation. AICD of T cell is characterized by the upregulation of death receptors and low persistence of T cells. Understanding the mechanism of AICD is crucial to improve the anti-tumor effect of CAR-T cells against solid tumors. Many approaches have been applied in CAR-T cell modification to enhance their anti-apoptosis ability. In this review, we summarized the molecular mechanisms of AICD in CAR-T cells and the progresses of anti-AICD in CAR-T cells therapy.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Morte Celular , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Imunoterapia Adotiva , Neoplasias/terapia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo
13.
Cancers (Basel) ; 14(19)2022 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-36230871

RESUMO

Bi-specific T cell engager (BiTE), an artificial bi-functional fusion protein, has shown promising therapeutic potential in preclinical and clinical studies. However, T cells cannot be sufficiently activated by BiTE, most likely due to lacking co-stimulatory signal. We reasoned that incorporating co-stimulatory signal might have the potential to enhance the T cell activation mediated by BiTE. We, therefore, designed a chimeric fusion protein, named as CD3εζ28, which consists of the CD3ε extracellular region, the CD28 costimulatory signal and the intracellular region of CD3ζ in tandem. T cells genetically modified to express both CD3εζ28 and GFP (T-CD3εζ28-GFP) were generated by retroviral transduction. The results from in vitro experiments showed that T-CD3εζCD28-GFP cells had superior cytotoxic effects on tumor cells in presence of BiTE compared with control T cells, as evidenced by IL-2 and IFN-γ production, T cell proliferation and sequential killing assay. In vivo, T-CD3εζCD28-GFP cells showed superior anti-tumor effects in Hela-BiTE. EGFRvIII xenograft tumor model, as evaluated by tumor growth rate and T cell persistence in comparison with control T cells. In order to further confirm these findings, we generated T cells modified to express both CD3εζCD28 on cell surface and BiTE.CD19 by autocrine manner (T-CD3εζCD28-BiTE.19). The superior anti-tumor effects of T-CD3εζCD28-BiTE.19 cells could also be evidenced by the similar in vitro and in vivo experiments; thus, incorporating co-stimulatory signal may be an effective approach to improve the effector function of T cells mediated by BiTE.

14.
Cancers (Basel) ; 14(15)2022 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-35954346

RESUMO

Elastin-like polypeptides (ELPs) are stimulus-responsive artificially designed proteins synthesized from the core amino acid sequence of human tropoelastin. ELPs have good biocompatibility and biodegradability and do not systemically induce adverse immune responses, making them a suitable module for drug delivery. Design strategies can equip ELPs with the ability to respond to changes in temperature and pH or the capacity to self-assemble into nanoparticles. These unique tunable biophysicochemical properties make ELPs among the most widely studied biopolymers employed in protein purification, drug delivery, tissue engineering and even in tumor therapy. As a module for drug delivery and as a carrier to target tumor cells, the combination of ELPs with therapeutic drugs, antibodies and photo-oxidation molecules has been shown to result in improved pharmacokinetic properties (prolonged half-life, drug targeting, cell penetration and controlled release) while restricting the cytotoxicity of the drug to a confined infected site. In this review, we summarize the latest developments in the application methods of ELP employed in tumor therapy, with a focus on its conjugation with peptide drugs, antibodies and photosensitizers.

15.
Mol Biol Rep ; 38(3): 1853-60, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20852941

RESUMO

Lactate dehydrogenase (LDH) catalyzes the reduction of pyruvate into lactate and constitutes a major checkpoint of anaerobic glycolysis. Recently, LDH draws a great deal of attention for its potential to be used as a novel diagnostic and therapeutic target for various diseases, including cancer and malaria. Insect LDHs have been mainly identified from fruit fly and mosquitoes, but not from silkworm. In this study, a novel LDH homologue, designated as BmLDH1, was firstly identified and characterized from the silkworm, Bombyx mori. The BmLDH1 cDNA contains an open reading frame of 996 bp, and encodes a protein of 331 amino acid residues with calculated molecular mass of 36 kDa. Sequence comparison showed BmLDH1 is a highly conserved protein. RT-PCR revealed BmLDH1 is transcripted in all tissues and in all developmental stages tested, indicating its essential roles for silkworm physiology and development. The BmLDH1 gene was subcloned and expressed in E. coli, and was further characterized by Western blot and Mass Spectrometry. The expressed protein contained the LDH activity, and could be inhibited by reduced glutathione in vitro. Immunofluoresence showed that the BmLDH1 was located in the cytoplasm. The cloned BmLDH1 sequence was deposited in the GenBank (accession number EU334850).


Assuntos
Bombyx/enzimologia , Bombyx/genética , Genes de Insetos/genética , L-Lactato Desidrogenase/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Bombyx/efeitos dos fármacos , Clonagem Molecular , Biologia Computacional , Ensaios Enzimáticos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glutationa/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Desidrogenase/antagonistas & inibidores , L-Lactato Desidrogenase/química , L-Lactato Desidrogenase/metabolismo , Dados de Sequência Molecular , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Transcrição Gênica/efeitos dos fármacos
16.
Toxicol Mech Methods ; 21(6): 444-52, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21466417

RESUMO

Comparative proteomic analysis was performed to identify proteins in the midgut of Takifugu rubripes (Fugu) in response to excessive fluoride. Sixteen fish were randomly divided into a control group and an experimental group. The control group was raised in soft water alone (F⁻= 0.4 mg/L), whereas the experimental group was raised in the soft water with sodium fluoride at a high concentration of 35 mg/L. After 3 days, proteins were extracted from the fish midgut and then subjected to two-dimensional (2-D) PAGE analysis. The matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI TOF/TOF MS) was applied to identify the differential expressed proteins between the two groups. Among 377 and 528 proteins detected in the control and the treated groups, respectively, 17 proteins were up-regulated and 218 were down-regulated (P < 0.01) in the fluoride-treated group, compared with the control group. We further analyzed 17 up-regulated proteins by MALDI TOF/TOF MS and identified 12 of them by MASCOT, of which eight were known proteins. Consistent with their annotated functions, these proteins seem to be involved in apoptosis and other functions related to fluorosis. Our results provide initial insights into the effects of excessive fluoride exposure on physiological and biochemical functions of Fugu midgut as well as on the toxicological mechanism of fluoride in both fish and human.


Assuntos
Proteínas de Peixes/metabolismo , Intoxicação por Flúor/metabolismo , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Bases de Dados de Proteínas , Regulação para Baixo/efeitos dos fármacos , Eletroforese em Gel Bidimensional , Proteínas de Peixes/química , Fluorose Dentária/metabolismo , Processamento de Imagem Assistida por Computador , Masculino , Mapeamento de Peptídeos , Proteômica/métodos , Distribuição Aleatória , Fluoreto de Sódio/administração & dosagem , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Takifugu , Espectrometria de Massas em Tandem , Regulação para Cima/efeitos dos fármacos
18.
Tumori ; 107(4): 341-352, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32988314

RESUMO

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) is an overexpressed antigen in esophageal squamous cell carcinomas (ESCCs) but with limited expression levels in normal esophageal tissues. Therefore, employing the adoptive transfer of T cells genetically modified to express chimeric antigen receptor (CAR) targeting HER2 could be a promising therapeutic strategy against ESCC. METHODS: Two different second-generation CAR-T cells expressing antibodies for HER2 and CD19 antigens were developed using retroviral vector transduction. The expression of HER2 antigen in ESCC tissue and cell lines was examined by immunohistochemistry and flow cytometry, respectively. The tumor killing efficacy of the CAR-T cells in mice model and ESCC cell lines and its potential for the treatment of ESCC was evaluated by determining tumor size in mice xenograft, and by crystal violet staining, MTS assay, and cytokine release. RESULTS: In vitro, HER2.CAR-T cells efficiently recognized and killed HER2-positive tumor cells as evidenced by the secretion of proinflammatory cytokines, interferon-γ, and interleukin 2 and by cytotoxicity assays. In vivo, intratumor injection of HER2.CAR-T cells resulted in a significant suppression of established ESCCs in a subcutaneous xenograft BALB/c nude mouse model. In contrast, the injection of CD19.CAR-T cells did not affect the tumor growth pattern. CONCLUSIONS: An effective HER2 CAR targeting ESCC was developed successfully. The HER2.CAR-T cell showed promising immunotherapeutic potential for the treatment of HER2-positive esophageal cancer.


Assuntos
Neoplasias Esofágicas/terapia , Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T/imunologia , Animais , Apoptose , Proliferação de Células , Técnicas de Cocultura , Neoplasias Esofágicas/imunologia , Neoplasias Esofágicas/patologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptor ErbB-2/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Am J Cancer Res ; 11(6): 2537-2567, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34249415

RESUMO

A majority of breast cancer patients die of widespread aggressive multidrug-resistant tumors. Aspartate ß-hydroxylase (ASPH) is an α-ketoglutarate-dependent dioxygenase and oncofetal antigen involved in embryogenesis. To illustrate if ASPH could be targeted for metastatic breast cancer, embedded and on-top three-dimensional (3-D) cultures, 3-D invasion, mammosphere formation, immunofluorescence, immunohistochemistry, Western blot, co-IP and microarray were conducted. In vitro metastasis was developed to imitate how cancer cells invade basement membrane at the primary site, transendothelially migrate, consequently colonize and outgrow at distant sites. Orthotopic and experimental pulmonary metastatic (tail vein injection) murine models were established using stable breast cancer cell lines. Cox proportional hazards regression models and Kaplan-Meier plots were applied to assess clinical outcome of breast cancer patients. In adult non-cancerous breast tissue, ASPH is undetectable. Pathologically, ASPH expression re-emerged at ductal carcinoma in situ (DCIS), and enhanced with disease progression, from early-stage invasive ductal carcinoma (IDC) to late-stage carcinoma. ASPH at moderate to high levels contribute to aggressive molecular subtypes, early relapse or more frequent progression and metastases, whereas substantially shortened overall survival and disease-free survival of breast cancer patients. Through direct physical interactions with A disintegrin and metalloproteinase domain-containing protein (ADAM)-12/ADAM-15, ASPH could activate SRC cascade, thus upregulating downstream components attributed to multifaceted metastasis. ASPH-SRC axis initiated pro-invasive invadopodium formation causing breakdown/disorganization of extracellular matrix (ECM), simultaneously potentiated epithelial-mesenchymal transition (EMT), induced cancer stem cell markers (CD44 and EpCAM), enhanced mammosphere formation and intensified 3-dimentional invasion. Oncogenic SRC upregulated matrix metallopeptidases (MMPs) were assembled by invadopodia, acting as executive effectors for multi-step metastasis. ASPH-SRC signal guided multi-organ metastases (to lungs, liver, bone, spleen, lymph nodes, mesentery or colon) in immunocompromised mice. Malignant phenotypes induced by ASPH-SRC axis were reversed by the third-generation small molecule inhibitor (SMI) specifically against ß-hydroxylase activity of ASPH in pre-clinical models of metastatic breast cancer. Collectively, ASPH could activate ADAMs-SRC-MMPs cascades to promote breast cancer tumor progression and metastasis. ASPH could direct invadopodium construction as a biomechanical sensor and pro-metastatic outlet. ASPH-mediated cancer progression could be specifically/efficiently subverted by SMIs of ß-hydroxylase activity. Therefore, ASPH emerges as a therapeutic target for breast cancer.

20.
Mol Biol Rep ; 37(6): 2903-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19821057

RESUMO

Ghrelin is an endogenous ligand for the growth hormone secretagogue receptor. It plays an important role in stimulating growth hormone secretion, food intake, body weight gain and gastric motility. cDNA sequences coding for ghrelin precursor protein (prepro-ghrelin) were isolated from the stomach of a giant panda. Two different mRNA sequences of ghrelin were obtained. The long open reading frame of ghrelin (354 bp) encodes a precursor protein of 117 amino acids with a 23 amino acid signal peptide. The short one (351 bp) encodes a precursor protein of 116 amino acids with the same 23 amino acid signal peptide. The presumed giant panda mature ghrelin proteins also had two forms. Comparative analysis showed that the first and the fourth amino acids (Gly and Phe) were completely conserved and the third amino acid (Ser) was also highly conserved in the mature ghrelin. RT-PCR analysis of giant panda ghrelin mRNA in various tissues revealed high level of expression in stomach, relative lower levels of expression in small intestine, liver and kidney, and no expression in thymus, spleen and heart.


Assuntos
DNA Complementar/genética , Regulação da Expressão Gênica , Grelina/genética , Ursidae/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Perfilação da Expressão Gênica , Grelina/química , Grelina/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa