Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 751
Filtrar
1.
Int J Mol Sci ; 24(23)2023 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-38069378

RESUMO

Patients receiving cranial radiotherapy for primary and metastatic brain tumors may experience radiation-induced brain injury (RIBI). Thus far, there has been a lack of effective preventive and therapeutic strategies for RIBI. Due to its complicated underlying pathogenic mechanisms, it is rather difficult to develop a single approach to target them simultaneously. We have recently reported that Reprimo (RPRM), a tumor suppressor gene, is a critical player in DNA damage repair, and RPRM deletion significantly confers radioresistance to mice. Herein, by using an RPRM knockout (KO) mouse model established in our laboratory, we found that RPRM deletion alleviated RIBI in mice via targeting its multiple underlying mechanisms. Specifically, RPRM knockout significantly reduced hippocampal DNA damage and apoptosis shortly after mice were exposed to whole-brain irradiation (WBI). For the late-delayed effect of WBI, RPRM knockout obviously ameliorated a radiation-induced decline in neurocognitive function and dramatically diminished WBI-induced neurogenesis inhibition. Moreover, RPRM KO mice exhibited a significantly lower level of acute and chronic inflammation response and microglial activation than wild-type (WT) mice post-WBI. Finally, we uncovered that RPRM knockout not only protected microglia against radiation-induced damage, thus preventing microglial activation, but also protected neurons and decreased the induction of CCL2 in neurons after irradiation, in turn attenuating the activation of microglial cells nearby through paracrine CCL2. Taken together, our results indicate that RPRM plays a crucial role in the occurrence of RIBI, suggesting that RPRM may serve as a novel potential target for the prevention and treatment of RIBI.


Assuntos
Lesões Encefálicas , Lesões por Radiação , Animais , Humanos , Camundongos , Apoptose , Encéfalo/patologia , Lesões Encefálicas/genética , Lesões Encefálicas/prevenção & controle , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/metabolismo , Inflamação/patologia , Microglia , Lesões por Radiação/genética , Lesões por Radiação/prevenção & controle , Lesões por Radiação/patologia
2.
J Med Chem ; 64(19): 14332-14343, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34524803

RESUMO

In addition to a variety of viral-glycoprotein receptors (e.g., heparan sulfate, Niemann-Pick C1, etc.), dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), from the C-type lectin receptor family, plays one of the most important pathogenic functions for a wide range of viruses (e.g., Ebola, human cytomegalovirus (HCMV), HIV-1, severe acute respiratory syndrome coronavirus 2, etc.) that invade host cells before replication; thus, its inhibition represents a relevant extracellular antiviral therapy. We report two novel p-tBu-calixarene glycoclusters 1 and 2, bearing tetrahydroxamic acid groups, which exhibit micromolar inhibition of soluble DC-SIGN binding and provide nanomolar IC50 inhibition of both DC-SIGN-dependent Jurkat cis-cell infection by viral particle pseudotyped with Ebola virus glycoprotein and the HCMV-gB-recombinant glycoprotein interaction with monocyte-derived dendritic cells expressing DC-SIGN. A unique cooperative involvement of sugar, linker, and calixarene core is likely behind the strong avidity of DC-SIGN for these low-valent systems. We claim herein new promising candidates for the rational development of a large spectrum of antiviral therapeutics.


Assuntos
Calixarenos/química , Moléculas de Adesão Celular/antagonistas & inibidores , Glicoconjugados/metabolismo , Glicoproteínas/antagonistas & inibidores , Ácidos Hidroxâmicos/química , Lectinas Tipo C/antagonistas & inibidores , Fenóis/química , Receptores de Superfície Celular/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Citomegalovirus/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Ebolavirus/fisiologia , Glicoconjugados/química , Glicoconjugados/farmacologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Células Jurkat , Lectinas Tipo C/metabolismo , Modelos Biológicos , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Virais/genética , Proteínas Virais/metabolismo
3.
J Immunol Res ; 2021: 5545173, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34250096

RESUMO

Fel d1 is an important allergen produced by cats that causes IgE reactions in up to 95% of cat-allergic adults. Immunotherapy to reduce human allergy to cats has demonstrated that people have the capacity to produce allergen-specific neutralizing antibodies that block IgE-mediated allergic responses. We wished to determine if "blocking" antibodies could be used to reduce the IgE binding ability of cat allergens prior to their exposure to humans. Here, we describe the characterization of Fel d1-specific antibodies. We demonstrated the efficacy of a rabbit polyclonal and an allergen-specific chicken IgY to bind to Fel d1 in cat saliva and block Fel d1-IgE binding and IgE-mediated basophil degranulation. Fel d1 blocking antibodies offer a new and exciting approach to the neutralization of cat allergens.


Assuntos
Alérgenos/imunologia , Anticorpos Bloqueadores/farmacologia , Glicoproteínas/antagonistas & inibidores , Hipersensibilidade Imediata/prevenção & controle , Animais de Estimação/imunologia , Animais , Anticorpos Bloqueadores/isolamento & purificação , Anticorpos Bloqueadores/uso terapêutico , Basófilos/efeitos dos fármacos , Basófilos/imunologia , Gatos , Degranulação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Galinhas , Glicoproteínas/imunologia , Humanos , Hipersensibilidade Imediata/imunologia , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Coelhos , Ratos , Saliva/imunologia
4.
Am J Physiol Cell Physiol ; 321(3): C415-C428, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34260299

RESUMO

Leucine-rich α-2-glycoprotein-1 (LRG1) is a novel profibrotic factor that modulates transforming growth factor-ß (TGF-ß) signaling. However, its role in the corneal fibrotic response remains unknown. In the present study, we found that the LRG1 level increased in alkali-burned mouse corneas. In the LRG1-treated alkali-burned corneas, there were higher fibrogenic protein expression and neutrophil infiltration. LRG1 promoted neutrophil chemotaxis and CXCL-1 secretion. Conversely, LRG1-specific siRNA reduced fibrogenic protein expression and neutrophil infiltration in the alkali-burned corneas. The clearance of neutrophils effectively attenuated the LRG1-enhanced corneal fibrotic response, whereas the presence of neutrophils enhanced the effect of LRG1 on the fibrotic response in cultured TKE2 cells. In addition, the topical application of LRG1 elevated interleukin-6 (IL-6) and p-Stat3 levels in the corneal epithelium and in isolated neutrophils. The clearance of neutrophils inhibited the expression of p-Stat3 and IL-6 promoted by LRG1 in alkali-burned corneas. Moreover, neutrophils significantly increased the production of IL-6 and p-Stat3 promoted by LRG1 in TKE2 cells. Furthermore, the inhibition of Stat3 signaling by S3I-201 decreased neutrophil infiltration and alleviated the LRG1-enhanced corneal fibrotic response in the alkali-burned corneas. S3I-201 also reduced LRG1 or neutrophil-induced fibrotic response in TKE2 cells. In conclusion, LRG1 promotes the corneal fibrotic response by stimulating neutrophil infiltration via the modulation of the IL-6/Stat3 signaling pathway. Therefore, LRG1 could be targeted as a promising therapeutic strategy for patients with corneal fibrosis.


Assuntos
Queimaduras Químicas/genética , Quimiotaxia/efeitos dos fármacos , Queimaduras Oculares/genética , Glicoproteínas/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética , Álcalis , Ácidos Aminossalicílicos/farmacologia , Animais , Benzenossulfonatos/farmacologia , Queimaduras Químicas/tratamento farmacológico , Queimaduras Químicas/metabolismo , Queimaduras Químicas/patologia , Linhagem Celular , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/metabolismo , Epitélio Corneano/patologia , Queimaduras Oculares/induzido quimicamente , Queimaduras Oculares/tratamento farmacológico , Queimaduras Oculares/patologia , Fibrose/prevenção & controle , Regulação da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
5.
Biophys Chem ; 270: 106537, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33450550

RESUMO

Nipah virus (NiV) infections are highly contagious and can cause severe febrile encephalitis. An outbreak of NiV infection has reported high mortality rates in Southeast Asian countries including Bangladesh, East Timor, Malaysia, Papua New Guinea, Vietnam, Cambodia, Indonesia, Madagascar, Philippines, Thailand and India. Considering the high risk for an epidemic outbreak, the World Health Organization (WHO) declared NiV as an emerging priority pathogen. However, there are no effective therapeutics or any FDA approved drugs available for the treatment of this infection. Among the known nine proteins of NiV, glycoprotein plays an important role in initiating the entry of viruses and attaching to the host cell receptors. Herein, three antiviral databases consisting of 79,892 chemical entities have been computationally screened against NiV glycoprotein (NiV-G). Particularly, multi-step molecular docking followed by extensive molecular binding interactions analyses, binding free energy estimation, in silico pharmacokinetics, synthetic accessibility and toxicity profile evaluations have been carried out for initial identification of potential NiV-G inhibitors. Further, molecular dynamics (MD) simulation has been performed to understand the dynamic properties of NiV-G protein-bound with proposed five inhibitors (G1-G5) and their interactions behavior, and any conformational changes in NiV-G protein during simulations. Moreover, Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) based binding free energies (∆G) has been calculated from all MD simulation trajectories to understand the energy contribution of each proposed compound in maintaining and stabilizing the complex binding interactions with NiV-G protein. Proposed compounds showed high negative ∆G values ranging from -166.246 to -226.652 kJ/mol indicating a strong affinity towards the NiV-G protein.


Assuntos
Antivirais/farmacologia , Glicoproteínas/antagonistas & inibidores , Vírus Nipah/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Virais/antagonistas & inibidores , Antivirais/química , Descoberta de Drogas , Glicoproteínas/química , Glicoproteínas/metabolismo , Infecções por Henipavirus/tratamento farmacológico , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Vírus Nipah/fisiologia , Bibliotecas de Moléculas Pequenas/química , Proteínas Virais/química , Proteínas Virais/metabolismo
6.
mBio ; 11(5)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934085

RESUMO

Membrane-associated RING-CH-type 8 (MARCH8) strongly blocks human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) incorporation into virions by downregulating its cell surface expression, but the mechanism is still unclear. We now report that MARCH8 also blocks the Ebola virus (EBOV) glycoprotein (GP) incorporation via surface downregulation. To understand how these viral fusion proteins are downregulated, we investigated the effects of MARCH8 on EBOV GP maturation and externalization via the conventional secretion pathway. MARCH8 interacted with EBOV GP and furin when detected by immunoprecipitation and retained the GP/furin complex in the Golgi when their location was tracked by a bimolecular fluorescence complementation (BiFC) assay. MARCH8 did not reduce the GP expression or affect the GP modification by high-mannose N-glycans in the endoplasmic reticulum (ER), but it inhibited the formation of complex N-glycans on the GP in the Golgi. Additionally, the GP O-glycosylation and furin-mediated proteolytic cleavage were also inhibited. Moreover, we identified a novel furin cleavage site on EBOV GP and found that only those fully glycosylated GPs were processed by furin and incorporated into virions. Furthermore, the GP shedding and secretion were all blocked by MARCH8. MARCH8 also blocked the furin-mediated cleavage of HIV-1 Env (gp160) and the highly pathogenic avian influenza virus H5N1 hemagglutinin (HA). We conclude that MARCH8 has a very broad antiviral activity by prohibiting different viral fusion proteins from glycosylation and proteolytic cleavage in the Golgi, which inhibits their transport from the Golgi to the plasma membrane and incorporation into virions.IMPORTANCE Enveloped viruses express three classes of fusion proteins that are required for their entry into host cells via mediating virus and cell membrane fusion. Class I fusion proteins are produced from influenza viruses, retroviruses, Ebola viruses, and coronaviruses. They are first synthesized as a type I transmembrane polypeptide precursor that is subsequently glycosylated and oligomerized. Most of these precursors are cleaved en route to the plasma membrane by a cellular protease furin in the late secretory pathway, generating the trimeric N-terminal receptor-binding and C-terminal fusion subunits. Here, we show that a cellular protein, MARCH8, specifically inhibits the furin-mediated cleavage of EBOV GP, HIV-1 Env, and H5N1 HA. Further analyses uncovered that MARCH8 blocked the EBOV GP glycosylation in the Golgi and inhibited its transport from the Golgi to the plasma membrane. Thus, MARCH8 has a very broad antiviral activity by specifically inactivating different viral fusion proteins.


Assuntos
Ebolavirus/química , Glicoproteínas/antagonistas & inibidores , HIV-1/química , Hemaglutininas Virais/metabolismo , Virus da Influenza A Subtipo H5N1/química , Ubiquitina-Proteína Ligases/genética , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Ebolavirus/fisiologia , Glicosilação , Células HEK293 , HIV-1/fisiologia , Células HeLa , Células Hep G2 , Humanos , Virus da Influenza A Subtipo H5N1/fisiologia , Ligação Proteica , Células THP-1 , Ubiquitina-Proteína Ligases/metabolismo , Células Vero , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/metabolismo
7.
Exp Cell Res ; 396(1): 112295, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32971116

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a fatal fibrosing interstitial lung disease with limited therapeutic options and a median survival of 3 years after diagnosis. Dysregulated epithelial regeneration is key event involved in initiating and sustaining IPF. The type II alveolar epithelial cells (AECIIs) play a crucial role for epithelial regeneration and stabilisation of alveoli. Loss of cell apical-basal polarity contributes to fibrosis. AECII has apical-basal polarity, but it is poorly understood whether AECII apical-basal polarity loss is involved in fibrosis. Bleomycin is a traditional inducer of pulmonary fibrosis. Here firstly we observed that bleomycin induced apical-basal polarity loss in cultured AECIIs. Next, cell polarity proteins lethal (2) giant larvae 1 (Lgl1), PAR-3A, aPKC and PAR-6B were investigated. We found bleomycin induced increases of Lgl1 protein and decreases of PAR-3A protein, and bleomycin-induced PAR-3A depression was mediated by increased-Lgl1. Then Lgl1 siRNA was transfected into AECIIs. Lgl1 siRNA prevented apical-basal polarity loss in bleomycin-treated AECIIs. At last, Lgl1-conditional knockout mice were applied in making animal models. Bleomycin induced pulmonary fibrosis, but this was attenuated in Lgl1-conditional knockout mice. Together, these data indicated that bleomycin mediated AECII apical-basal polarity loss which contributed to experimental pulmonary fibrosis. Inhibition of Lgl1 should be a potential therapeutic strategy for the disease.


Assuntos
Células Epiteliais Alveolares/efeitos dos fármacos , Bleomicina/farmacologia , Polaridade Celular/efeitos dos fármacos , Glicoproteínas/genética , Fibrose Pulmonar/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Polaridade Celular/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/metabolismo , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos Knockout , Cultura Primária de Células , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Fibrose Pulmonar/prevenção & controle , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais
8.
Antiviral Res ; 174: 104701, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31877348

RESUMO

Lassa virus (LASV) is the causative agent of Lassa hemorrhagic fever in humans, and the limited therapeutic treatment for Lassa fever poses significant threat to public health in West Africa. Using an HIV based pseudovirus platform, we identified isavuconazole, a triazole antifungal for systemic use, as a LASV entry inhibitor with an EC50 of 1.2 µM. Isavuconazole inhibits Lassa virus entry by blocking the pH dependent viral fusion mediated by the Lassa virus surface glycoprotein. Fragment replacement mutational study indicated that isavuconazole targets the stable signal peptide (SSP)-membrane fusion subunit (GP2) interface of Lassa glycoprotein. Further mutational study of the SSP-GP2 region of LASV glycoprotein revealed that S27 in the N-terminal transmembrane region of SSP and V431, F434 and V435 in the transmembrane domain of GP2 affect anti-LASV activity of isavuconazole. Isavuconazole also displays antiviral activity to five New World (NW) mammarenaviruses that cause hemorrhagic fever. This study facilitates the potential repurposing of isavuconazole for therapeutic intervention against human-pathogenic arenaviruses, and provides the basis for further structural optimization of arenavirus fusion inhibitors based on the predicted structural characteristics of the unique SSP-GP2 interface.


Assuntos
Antivirais/farmacologia , Glicoproteínas/antagonistas & inibidores , Vírus Lassa/efeitos dos fármacos , Nitrilas/farmacologia , Piridinas/farmacologia , Triazóis/farmacologia , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Internalização do Vírus/efeitos dos fármacos , Células A549 , Antifúngicos/farmacologia , Linhagem Celular , Reposicionamento de Medicamentos , Células HEK293 , Humanos , Febre Lassa/tratamento farmacológico , Vírus Lassa/química , Sinais Direcionadores de Proteínas
9.
Eur Rev Med Pharmacol Sci ; 23(20): 8870-8877, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31696489

RESUMO

OBJECTIVE: MiR-1 has been reported to act as an inhibitory microRNA in gastric cancer (GC). This study aimed to investigate the regulatory mechanism by which miR-1-3p blocks the progression of GC by targeting stanniocalcin 2 (STC2). PATIENTS AND METHODS: The expression level of miR-1-3p in GC was assessed via quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). Expressions of STC2 were measured by qRT-PCR and Western blot analysis. Proliferation and invasion assays were detected by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) and transwell assays, respectively. Moreover, the dual-luciferase reporter assay was used to confirm the binding sites between miR-1-3p and STC2. RESULTS: MiR-1-3p was significantly down-regulated in GC. Moreover, abnormal expression of miR-1-3p was correlated with GC tumor size. Functionally, overexpression of miR-1-3p inhibited proliferation and invasion in GC by inhibiting stanniocalcin 2 (STC2) expressions. In contrast, STC2 was significantly up-regulated in GC. Furthermore, miR-1-3p negatively regulated STC2 expression in GC. The upregulation of STC2 weakened the inhibitory effect of miR-1-3p in GC. CONCLUSIONS: MiR-1-3p suppressed cell proliferation and invasion by targeting STC2 in GC, providing a novel therapeutic target for GC.


Assuntos
Proliferação de Células , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , MicroRNAs/metabolismo , Neoplasias Gástricas/patologia , Regiões 3' não Traduzidas , Antagomirs/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Pessoa de Meia-Idade , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência , Neoplasias Gástricas/genética
10.
Cell Signal ; 63: 109381, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31374291

RESUMO

Multidrug resistance is a major treatment obstacle for recurrent and metastatic bladder cancer, which often leads to disease progression and poor clinical outcome. Although overexpression of interleukin-6 (IL-6) appears to play a critical role in the development of chemotherapy resistance, inhibitors for IL-6 alone have not improved clinical outcomes. Since the IL-6/IL-6R/GP130 complex is involved in multidrug resistance, another strategy would be to focus on glycoprotein-130 (GP130) since it dimerizes with IL-6R/CD26 as a membrane-bound signaling transducer receptor and initiates subsequent signaling activation and may be a potential therapeutic target. Currently, the role of GP130 in chemoresistant bladder cancer is unknown. In the present study, we demonstrate that GP130 is over-expressed in cisplatin and gemcitabine-resistant bladder cancer cells, and that the inhibition of GP130 expression significantly reduces cell viability, survival and migration. Downstream of GP130 is PI3K/AKT/mTOR signaling, which is inactivated by SC144, a GP130 inhibitor. However, Raf/MEK/ERK signaling, which also is downstream of GP130 is activated by SC144. This activation is likely based on a mTOR/S6K1/PI3K/ERK negative feedback loop, which is presumed to counteract the inhibitory effect of SC144 on tumor aggressiveness. Blocking both GP130 and pERK resulted in synergistic inhibition of cytotoxicity, clonal survival rates and cell migration in our chemotherapy resistant bladder cancer cells. This vertical inhibition offers a novel therapeutic strategy for targeting human chemoresistant bladder cancer.


Assuntos
Carcinoma de Células de Transição/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glicoproteínas/antagonistas & inibidores , Hidrazinas/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Quinoxalinas/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Butadienos/farmacologia , Butadienos/uso terapêutico , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Neoplasias da Bexiga Urinária/patologia
11.
Biofactors ; 45(6): 944-949, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31348577

RESUMO

Dermal-epidermal interaction plays a role in many pathophysiological processes, such as tumor invasion and psoriasis, as well as wound healing, and is mediated at least in part by secretory factors. In this study, we investigated the factor(s) involved. We found that stanniocalcin-1 (STC1), a cytokine, is expressed at the basal layer of epidermis. Knockdown of STC1 with siRNA in HaCaT cells decreased matrix metalloproteinase 1 (MMP1) expression, suggesting that STC1 serves as an autocrine factor, maintaining MMP1 mRNA expression in the epidermal layer. In dermal fibroblasts, STC1 increased MMP1 mRNA expression and decreased collagen1A1 and elastin mRNA expression. These actions were inhibited by SP600125, a jun kinase (JNK) inhibitor. Nuclear translocation of AP-1, a downstream signal of JNK, was implicated in the actions of STC1. In a coculture system of HaCaT cells and fibroblasts, used as a model of dermal-epidermal interaction, knockdown of STC1 in HaCaT cells with siRNA reduced the negative effects (i.e., induction of MMP1 and decrease of collagen1A1 and elastin) of STC1 on fibroblasts. These results suggest that STC1 secreted from the epidermal layer is a mediator of dermal-epidermal interaction.


Assuntos
Epiderme/metabolismo , Fibroblastos/metabolismo , Glicoproteínas/genética , Metaloproteinase 1 da Matriz/genética , Comunicação Autócrina/genética , Linhagem Celular , Movimento Celular/genética , Técnicas de Cocultura , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Glicoproteínas/antagonistas & inibidores , Humanos , Fosforilação/genética , RNA Interferente Pequeno/genética , Fator de Transcrição AP-1/genética , Cicatrização/genética
12.
Cell Prolif ; 52(5): e12651, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31297902

RESUMO

OBJECTIVE: It is essential to characterize underlying molecular mechanism associated with head and neck squamous cell carcinoma (HNSCC) and identify promising therapeutic targets. Herein, we explored role of homeobox transcript antisense RNA (HOTAIR) in HNSCC to regulate stanniocalcin-2 (STC2) by sponging microRNA-206 (miR-206). METHODS: HNSCC-related differentially expressed genes and regulation network amongst HOTAIR, miR-206 and STC2 were identified. Next, effect of HOTAIR on cell biological functions of HNSCC was identified after transfection of cells with HOTAIR overexpressed plasmids or siRNA against HOTAIR. PI3K/AKT signalling pathway-related gene expression was measured after miR-206 and STC2 were suppressed. Cell invasion, migration and proliferation were assessed. Finally, tumour growth was assessed to determine the effects of HOTAIR/miR-206/STC2 axis in vivo. RESULTS: HOTAIR specifically bound to miR-206 and miR-206 targeted STC2. Downregulated HOTAIR or upregulated miR-206 suppressed HNSCC cell proliferation, invasion and migration. miR-206 inhibited PI3K/AKT signalling pathway by down-regulating STC2. Besides, silenced HOTAIR or overexpressed miR-206 repressed the tumour growth of nude mice with HNSCC. CONCLUSION: HOTAIR regulated HNSCC cell biological functions by binding to miR-206 through STC2.


Assuntos
Glicoproteínas/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Nus , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo
13.
Mol Med Rep ; 20(2): 977-984, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31173256

RESUMO

Stanniocalcin 2 (STC2), a secretory glycoprotein hormone, regulates many biological processes including cell proliferation, apoptosis, tumorigenesis and atherosclerosis. However, the effect of STC2 on proliferation, migration and epithelial­mesenchymal transition (EMT) progression in human colorectal cancer (CRC) cells remains poorly understood. The expression level of STC2 was determined by quantitative real­time polymerase chain reaction (qPCR) and western blot analysis. Cell Counting Kit­8 (CCK­8) was used to detect the viability of SW480 cells. The invasion and migration of cells were identified by wound healing and Transwell assays. The mRNA and protein expression levels of ß­catenin, matrix metalloproteinase (MMP)­2, MMP­9, E­cadherin and vimentin were assessed by qPCR and western blot analysis. In the present study, it was demonstrated that STC2 was highly expressed in the CRC cell lines. After silencing of STC2, the cell viability, migration and invasion were significantly reduced. Silencing of STC2 in the CRC Sw480 cells increased the expression of E­cadherin and decreased the expression of vimentin, MMP­2 and MMP­9, compared to those in the normal and empty vector group. Furthermore, the expression of ß­catenin in the STC2 gene silenced group was suppressed, and the expression of ß­catenin was reversed by Wnt activator, SB216763. These results demonstrated that STC2 participates in the development and progression of CRC by promoting CRC cell proliferation, survival and migration and activating the Wnt/ß­catenin signaling pathway.


Assuntos
Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , beta Catenina/genética , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/metabolismo , Células HCT116 , Células HT29 , Humanos , Indóis/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Maleimidas/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Vimentina/genética , Vimentina/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
14.
Life Sci ; 229: 261-276, 2019 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-31082400

RESUMO

AIM: Myokines are associated with regulation of bone and muscle mass. However, limited information is available regarding the impact of myokines on glucocorticoid (GC) mediated adverse effects on the musculoskeletal system. This study investigates the role of myokine fibroblast growth factor-2 (FGF-2) in regulating GC-induced deleterious effects on bone and skeletal muscle. METHODS: Primary osteoblast cells and C2C12 myoblast cell line were treated with FGF-2 and then exposed to dexamethasone (GC). FGF-2 mediated attenuation of the inhibitory effect of GC on osteoblast and myoblast differentiation and muscle atrophy was assessed through quantitative PCR and western blot analysis. Further, FGF-2 was administered subcutaneously to dexamethasone treated mice to collect bone and skeletal muscle tissue for in vivo analysis of bone microarchitecture, mechanical strength, histomorphometry and for histological alterations in treated tissue samples. KEY FINDINGS: FGF-2 abrogated the dexamethasone induced inhibitory effect on osteoblast differentiation by modulating BMP-2 pathway and inhibiting Wnt antagonist sclerostin. Further, dexamethasone induced atrophy in C2C12 cells was mitigated by FGF-2 as evident from down regulation of atrogenes expression. FGF-2 prevented GC-induced impairment of mineral density, biomechanical strength, trabecular bone volume, cortical thickness and bone formation rate in mice. Additionally, skeletal muscle tissue from GC treated mice displayed weak myostatin immunostaining and reduced expression of atrogenes following FGF-2 treatment. SIGNIFICANCE: FGF-2 mitigated GC induced effects through inhibition of sclerostin and myostatin expression in bone and muscle respectively. Taken together, this study exhibited the role of exogenous FGF-2 in sustaining osteoblastogenesis and inhibiting muscle atrophy in presence of glucocorticoid.


Assuntos
Osso e Ossos/metabolismo , Dexametasona/toxicidade , Fator 2 de Crescimento de Fibroblastos/farmacologia , Glicoproteínas/antagonistas & inibidores , Músculo Esquelético/metabolismo , Doenças Musculoesqueléticas/tratamento farmacológico , Miostatina/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Diferenciação Celular , Células Cultivadas , Glucocorticoides/toxicidade , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Doenças Musculoesqueléticas/induzido quimicamente , Doenças Musculoesqueléticas/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos
15.
Dev Comp Immunol ; 99: 103398, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31121186

RESUMO

Emerging evidence suggests that CD83, a dendritic cells (DCs) maturation marker in humans and mice, may prossess immunomodulatory capacities. Although porcine CD83 shares ∼75% sequence homology with its human counterpart, whether it functions as an immunoregulatory molecule remains unknown. To investigate porcine CD83 function, we deleted it in porcine DCs by RNA intereference. Results show that membrane-bound CD83 (mCD83) promotes DC-mediated T cell proliferation and cytokine production, thus confirming its immunoregulatory capacity. Intriguingly, porcine soluble CD83 (sCD83) treatment instead led to inhibition of DC-mediated T cell activation. Moreover, porcine sCD83 also inhibited differentiation of prepheral blood mononuclear cells (PBMCs) into DCs. These results collectively indicate that in addition to being a DC maturation maker, both membrane bound and souble porcine CD83 serve as immunoregulatory molecules with opposite effects on DC-mediated T cell activation and DC differentiation.


Assuntos
Antígenos CD/imunologia , Células Dendríticas/imunologia , Glicoproteínas/imunologia , Imunoglobulinas/imunologia , Glicoproteínas de Membrana/imunologia , Suínos/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD/genética , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/genética , Células HEK293 , Humanos , Imunoglobulinas/genética , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , RNA Interferente Pequeno , Antígeno CD83
16.
Oncol Rep ; 41(6): 3270-3280, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31002347

RESUMO

Leucine­rich­alpha­2­glycoprotein 1 (LRG­1) has been reported to be associated with multiple malignancies. However, its participation in thyroid carcinoma progression remains unclear. In the present study, the biological function and underlying molecular mechanisms of LRG­1 in thyroid carcinoma were investigated. It was found that LRG­1 was overexpressed in thyroid carcinoma tissues, and high LRG­1 expression predicted poor patient survival and late tumor stage. As shown in the mouse xenograft study, knockdown of LRG­1 significantly attenuated thyroid cancer growth in vivo. Based on wound healing, Transwell, proliferation and apoptosis assays, it was found that the knockdown of LRG­1, using shLRG­1, inhibited cell migration and invasion, but did not affect proliferation and apoptosis in thyroid cancer cells. Furthermore, LRG­1 also induced epithelial­mesenchymal transition (EMT) in thyroid carcinoma cells. Western blot analysis revealed that this tumor­promoting bioactivity of LRG­1 was attributed to its selective activation of MAPK/p38 signaling. All of these findings indicate that LRG­1 plays a deleterious role in the progression of thyroid carcinoma. LRG­1 may serve as a promising biomarker for predicting prognosis in thyroid carcinoma patients, and LRG­1­based therapy may be developed into a novel strategy for the treatment of thyroid carcinoma.


Assuntos
Biomarcadores Tumorais/genética , Transição Epitelial-Mesenquimal/genética , Glicoproteínas/genética , Neoplasias da Glândula Tireoide/genética , Adolescente , Adulto , Idoso , Animais , Apoptose/genética , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Glicoproteínas/antagonistas & inibidores , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Prognóstico , Transdução de Sinais , Neoplasias da Glândula Tireoide/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem , Proteínas Quinases p38 Ativadas por Mitógeno/genética
17.
J Cancer Res Ther ; 15(1): 148-152, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30880771

RESUMO

CONTEXT: Resistance of cancer cells to chemotherapeutic drugs is a major pitfall of the failure of chemotherapy treatment for cholangiocarcinoma (CCA). A new therapeutic strategy that can improve treatment efficacy is mandatory for CCA patients. Our previous findings demonstrated the overexpression of methionine aminopeptidase-2 (MetAP2) in CCA patients. In addition, supplementation of TNP-470, a MetAP2 inhibitor, significantly inhibited the growth and metastatic activities of CCA cell lines. However, the molecular mechanism of antitumor activity of TNP-470 and the synergistic antitumor activity of TNP-470 combined with chemotherapeutic drugs are still unknown. AIMS: The aim of this study is to evaluate the molecular mechanism of anticancer activity and the potential use of TNP-470 as a chemosensitizing agent in CCA cell lines. MATERIALS AND METHODS: Cell cycle and apoptosis of CCA cell lines were evaluated using flow cytometry with propidium iodide staining. Expression of apoptosis regulatory proteins was measured by Western blotting. The chemosensitizing effect of TNP-470 was determined using combination index. RESULTS: TNP-470 inhibited the growth of CCA cells via induction of apoptosis through activation of the p38-phosphorylation and up- and down-regulation of Bax and Bcl-xL, respectively. Furthermore, TNP-470 significantly enhanced the antitumor activity of 5-fluorouracil, cisplatin, doxorubicin, and gemcitabine. CONCLUSIONS: The present results show that TNP-470 could be a potential therapeutic or adjuvant agent for CCA.


Assuntos
Aminopeptidases/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias dos Ductos Biliares/tratamento farmacológico , Colangiocarcinoma/tratamento farmacológico , Glicoproteínas/antagonistas & inibidores , O-(Cloroacetilcarbamoil)fumagilol/farmacologia , Aminopeptidases/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias dos Ductos Biliares/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colangiocarcinoma/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Glicoproteínas/metabolismo , Humanos , Concentração Inibidora 50 , Metionil Aminopeptidases , O-(Cloroacetilcarbamoil)fumagilol/uso terapêutico
18.
Biol Trace Elem Res ; 192(2): 263-276, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30790121

RESUMO

Selenium (Se), as a nutritionally essential trace element, has been shown to decrease with age and is closely related to Alzheimer's disease (AD). To probe the effects of Se on AD pathology, two-dimensional fluorescence difference gel electrophoresis was applied to the serum samples collected from the wild-type (WT) mice and the triple transgenic (PS1M146V/AßPPSwe/TauP301L) AD mice (3xTg-AD), treated with or without sodium selenate in drinking water for 4 months beginning at 2 months of age. Proteomics results revealed 17 differentially expressed proteins between WT and 3xTg-AD mice. It was found that the administration of selenate reversed the alterations of the differentially expressed serum proteins by up-regulating 13 proteins and down-regulating 2 proteins which were reported to be involved in the key pathogenesis of AD, including regulation of Aß production, lipid metabolism regulation, and anti-inflammation. These results suggested that a dietary supplement with selenate is effective for prevention and treatment of AD, and the mechanism was maybe related to its role in Aß regulation, lipid metabolism, and anti-inflammation. Moreover, we also presented that α-2 macroglobulin, transthyretin, haptoglobin, alpha-2-HS-glycoprotein, and alpha-1-antitrypsin in the serum can be used to evaluate the effect of selenate on AD pathology.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Modelos Animais de Doenças , Proteômica , Ácido Selênico/farmacologia , Doença de Alzheimer/sangue , Doença de Alzheimer/patologia , Animais , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/sangue , Haptoglobinas/análise , Haptoglobinas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Pré-Albumina/análise , Pré-Albumina/antagonistas & inibidores , alfa 2-Macroglobulinas Associadas à Gravidez/análise , alfa 2-Macroglobulinas Associadas à Gravidez/antagonistas & inibidores , alfa 1-Antitripsina/sangue , alfa 1-Antitripsina/metabolismo
19.
J Cell Biochem ; 120(6): 10310-10322, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30556161

RESUMO

Extensive evidence indicate that long noncoding RNAs (lncRNAs) regulates the tumorigenesis and progression of hepatocellular carcinoma (HCC). However, the expression and biological function of lncRNA A1BG antisense RNA 1 (A1BG-AS1) were poorly known in HCC. Here, we found the underexpression of A1BG-AS1 in HCC via analysis of The Cancer Genome Atlas database. Further analyses confirmed that A1BG-AS1 expression in HCC was markedly lower than that in noncancerous tissues based on our HCC cohort. Clinical association analysis revealed that low A1BG-AS1 expression correlated with poor prognostic features, such as microvascular invasion, high tumor grade, and advanced tumor stage. Follow-up data indicated that low A1BG-AS1 level evidently correlated with poor clinical outcomes of HCC patients. Moreover, forced expression of A1BG-AS1 repressed proliferation, migration, and invasion of HCC cells in vitro. Conversely, A1BG-AS1 knockdown promoted these malignant behaviors in HepG2 cells. Mechanistically, A1BG-AS1 functioned as a competing endogenous RNA by directly sponging miR-216a-5p in HCC cells. Notably, miR-216a-5p restoration rescued A1BG-AS1 attenuated proliferation, migration and invasion of HCCLM3 cells. A1BG-AS1 positively regulated the levels of phosphatase and tensin homolog and SMAD family member 7, which were reduced by miR-216a-5p in HCC cells. Altogether, we conclude that A1BG-AS1 exerts a tumor suppressive role in HCC progression.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/patologia , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/antagonistas & inibidores , MicroRNAs/genética , RNA Longo não Codificante/genética , Apoptose , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Movimento Celular , Feminino , Seguimentos , Glicoproteínas/genética , Humanos , Imunoglobulinas/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Oligonucleotídeos Antissenso/genética , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Prognóstico , Proteína Smad7/genética , Proteína Smad7/metabolismo , Células Tumorais Cultivadas
20.
Front Immunol ; 9: 2268, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30510550

RESUMO

Impressive clinical efficacy of chimeric antigen receptor (CAR)-engineered T cell therapy for hematological malignancies have prompted significant efforts in achieving similar responses in solid tumors. The lack of truly restricted and uniform expression of tumor-associated antigens, as well as limited T cell persistence and/or tumor trafficking pose major challenges for successful translation of CAR T cell therapy in solid tumors. Recent studies have demonstrated that aberrantly glycosylated cell surface proteins on tumor cells are amenable CAR targets. Tumor-associated glycoprotein 72 (TAG72) antigen is the sialyl-Tn found on multiple O-glycoproteins expressed at high levels on the surface of several cancer types, including ovarian cancer. Here, we developed a humanized TAG72-specific CAR containing a 4-1BB intracellular co-stimulatory signaling domain (TAG72-BBζ). TAG72-BBζ CAR T cells showed potent antigen-dependent cytotoxicity and cytokine production against multiple TAG72+ ovarian cancer cell lines and patient-derived ovarian cancer ascites. Using in vivo xenograft models of peritoneal ovarian tumors, regional intraperitoneal delivery of TAG72-BBζ CAR T cells significantly reduced tumor growth, extended overall survival of mice, and was further improved with repeat infusions of CAR T cells. However, reduced TAG72 expression was observed in early recurring tumors, which coincided with a lack of T cell persistence. Taken together, we demonstrate efficacy with TAG72-CAR T cells in ovarian cancer, warranting further investigations as a CAR T cell therapeutic strategy for this disease.


Assuntos
Glicoproteínas/antagonistas & inibidores , Imunoterapia Adotiva/métodos , Neoplasias Ovarianas/terapia , Neoplasias Peritoneais/terapia , Receptores de Antígenos Quiméricos/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Feminino , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Humanos , Estimativa de Kaplan-Meier , Camundongos , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Neoplasias Peritoneais/imunologia , Neoplasias Peritoneais/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Carga Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA