Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Hum Vaccin Immunother ; 19(1): 2202127, 2023 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-37128699

RESUMEN

DNA vaccines containing only antigenic components have limited efficacy and may fail to induce effective immune responses. Consequently, adjuvant molecules are often added to enhance immunogenicity. In this study, we generated a tumor vaccine using a plasmid encoding NMM (NY-ESO-1/MAGE-A3/MUC1) target antigens and immune-associated molecules. The products of the vaccine were analyzed in 293 T cells by western blotting, flow cytometry, and meso-scale discovery electrochemiluminescence. To assess the immunogenicity obtained, C57BL/6 mice were immunized using the DNA vaccine. The results revealed that following immunization, this DNA vaccine induced cellular immune responses in C57BL/6 mice, as evaluated by the release of IFN-γ, and we also detected increases in the percentages of nonspecific lymphocytes, as well as those of antigen-specific T cells. Furthermore, immunization with the pNMM vaccine was found to significantly inhibit tumor growth and prolonged the survival of mice with B16-NMM+-tumors. Our data revealed that pNMM DNA vaccines not only confer enhanced immunity against tumors but also provide a potentially novel approach for vaccine design. Moreover, our findings provide a basis for further studies on vaccine pharmacodynamics and pharmacology, and lay a solid foundation for clinical application.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Vacunas de ADN , Ratones , Animales , Ratones Endogámicos C57BL , Antígenos de Neoplasias , Adyuvantes Inmunológicos , Inmunidad Celular
2.
Pharmaceutics ; 15(3)2023 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-36986633

RESUMEN

Numerous drugs have emerged to treat various diseases, such as COVID-19, cancer, and protect human health. Approximately 40% of them are lipophilic and are used for treating diseases through various delivery routes, including skin absorption, oral administration, and injection. However, as lipophilic drugs have a low solubility in the human body, drug delivery systems (DDSs) are being actively developed to increase drug bioavailability. Liposomes, micro-sponges, and polymer-based nanoparticles have been proposed as DDS carriers for lipophilic drugs. However, their instability, cytotoxicity, and lack of targeting ability limit their commercialization. Lipid nanoparticles (LNPs) have fewer side effects, excellent biocompatibility, and high physical stability. LNPs are considered efficient vehicles of lipophilic drugs owing to their lipid-based internal structure. In addition, recent LNP studies suggest that the bioavailability of LNP can be increased through surface modifications, such as PEGylation, chitosan, and surfactant protein coating. Thus, their combinations have an abundant utilization potential in the fields of DDSs for carrying lipophilic drugs. In this review, the functions and efficiencies of various types of LNPs and surface modifications developed to optimize lipophilic drug delivery are discussed.

3.
Medicine (Baltimore) ; 98(28): e16256, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31305407

RESUMEN

RATIONALE: Fabry's disease is an X-linked inherited syndrome. Herein, we presented an unusual case of Fabry disease coexisting with immunoglobulin A nephropathy (IgAN) presenting with Alport syndrome-like pathological findings. PATIENT CONCERNS: We report a 30-year-old male who presented with proteinuria and elevated serum creatinine and for whom the initial pathologic diagnosis supported Alport syndrome. DIAGNOSES: A diagnosis of Fabry disease with immunoglobulin A nephropathy (IgAN) was finally made after further examination. INTERVENTIONS: After the initial diagnosis the patient was treated with herbal medications and mecobalamin. OUTCOMES: The patient was discharged 1 week later. He was maintained on these treatments and received regular follow-up in our hospital. LESSONS SUBSECTIONS AS PER STYLE: FD coexisting with IgAN is rare and may have nonspecific clinical presentations. Laboratory examination and genetic diagnosis is needed for confirmation. Timely diagnosis and reproductive intervention is needed for therapy.


Asunto(s)
Enfermedad de Fabry/complicaciones , Enfermedad de Fabry/diagnóstico , Nefritis/complicaciones , Nefritis/diagnóstico , Adulto , Diagnóstico Diferencial , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/inmunología , Humanos , Inmunoglobulina A/metabolismo , Riñón/patología , Masculino , Nefritis/tratamiento farmacológico , Nefritis/inmunología
4.
J Cell Sci ; 132(10)2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31028177

RESUMEN

Necroptosis is a regulated form of necrotic cell death that is mediated by receptor-interacting serine/threonine-protein kinase 1 (RIPK1), RIPK3 and mixed-lineage kinase domain-like protein (MLKL), which mediates necroptotic signal transduction induced by tumor necrosis factor (TNF). Although many target proteins for necroptosis have been identified, no report had indicated that FK506-binding protein 12 (FKBP12, also known as FKBP1A), an endogenous protein that regulates protein folding and conformation alteration, is involved in mediating necroptosis. In this study, we found that FKBP12 acts as a novel target protein in mediating necroptosis and the related systemic inflammatory response syndrome triggered by TNF. The mechanistic study discovered that FKBP12 is essential for initiating necrosome formation and RIPK1-RIPK3-MLKL signaling pathway activation in response to TNF receptor 1 ligation. In addition, FKBP12 is indispensable for RIPK1 and RIPK3 expression and subsequent spontaneous phosphorylation, which are essential processes for initial necrosome formation and necroptotic signal transduction; therefore, FKBP12 may target RIPK1 and RIPK3 to mediate necroptosis in vitro and in vivo Collectively, our data demonstrate that FKBP12 could be a potential therapeutic target for the clinical treatment of necroptosis-associated diseases.


Asunto(s)
Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína 1A de Unión a Tacrolimus/metabolismo , Animales , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Necroptosis/fisiología , Fosforilación , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/patología , Serina-Treonina Quinasas TOR/metabolismo
5.
J Cell Mol Med ; 23(4): 2489-2504, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30672108

RESUMEN

As a nitric oxide (NO) donor prodrug, JS-K inhibits cancer cell proliferation, induces the differentiation of human leukaemia cells, and triggers apoptotic cell death in various cancer models. However, the anti-cancer effect of JS-K in gastric cancer has not been reported. In this study, we found that JS-K inhibited the proliferation of gastric cancer cells in vitro and in vivo and triggered mitochondrial apoptosis. Moreover, JS-K induced a significant accumulation of reactive oxygen species (ROS), and the clearance of ROS by antioxidant reagents reversed JS-K-induced toxicity in gastric cancer cells and subcutaneous xenografts. Although JS-K triggered significant NO release, NO scavenging had no effect on JS-K-induced toxicity in vivo and in vitro. Therefore, ROS, but not NO, mediated the anti-cancer effects of JS-K in gastric cancer. We also explored the potential mechanism of JS-K-induced ROS accumulation and found that JS-K significantly down-regulated the core proteins of mitochondria respiratory chain (MRC) complex I and IV, resulting in the reduction of MRC complex I and IV activity and the subsequent ROS production. Moreover, JS-K inhibited the expression of antioxidant enzymes, including copper-zinc-containing superoxide dismutase (SOD1) and catalase, which contributed to the decrease of antioxidant enzymes activity and the subsequent inhibition of ROS clearance. Therefore, JS-K may target MRC complex I and IV and antioxidant enzymes to exert ROS-dependent anti-cancer function, leading to the potential usage of JS-K in the prevention and treatment of gastric cancer.


Asunto(s)
Compuestos Azo/farmacología , Proliferación Celular/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Piperazinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Catalasa/genética , Línea Celular Tumoral , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/genética , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Óxido Nítrico/genética , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Superóxido Dismutasa-1/genética
6.
Front Cell Dev Biol ; 7: 393, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32039207

RESUMEN

As a programmed necrotic cell death, necroptosis has the intrinsic initiators, including receptor-interacting serine/threonine-protein kinase 1 (RIPK1), RIPK3 and mixed-lineage kinase domain-like protein (MLKL), which combine to form necroptotic signaling pathway and mediate necroptosis induced by various necroptotic stimuli, such as tumor necrosis factor (TNF). Although chemical inhibition of RIPK1 blocks TNF-induced necroptosis, genetic elimination of RIPK1 does not suppress but facilitate necroptosis triggered by TNF. Moreover, RIPK3 has been reported to mediate the RIPK1-independent necroptosis, but the involved mechanism is unclear. In this study, we found that TRADD was essential for TNF-induced necroptosis in RIPK1-knockdown L929 and HT-22 cells. Mechanistic study demonstrated that TRADD bound RIPK3 to form new protein complex, which then promoted RIPK3 phosphorylation via facilitating RIPK3 oligomerization, leading to RIPK3-MLKL signaling pathway activation. Therefore, TRADD acted as a partner of RIPK3 to initiate necroptosis in RIPK1-knockdown L929 and HT-22 cells in response to TNF stimulation. In addition, TRADD was critical for the accumulation of reactive oxygen species (ROS), which contributed to RIPK1-independent necroptosis triggered by TNF. Collectively, our data demonstrate that TRADD acts as the new target protein for TNF-induced RIPK3 activation and the subsequent necroptosis in a RIPK1-independent manner.

7.
Sci Rep ; 7(1): 16111, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29170425

RESUMEN

Receptor-interacting protein kinase 3 (RIP3) is a critical initiator in mediating necroptosis induced by tumor necrosis factor alpha (TNFα) in L929 cells, so knockdown of RIP3 inhibits TNFα-induced L929 cell necroptosis. However, RIP3 knockdown was shown to switch TNFα-induced necroptosis to apoptosis in L929 cells in other studies. Therefore, whether RIP3 knockdown blocks the TNFα-induced death of L929 cells is controversial. In this study, TNFα activated caspase pathway and induced cell death in RIP3 knockdown L929 cells, and the RIP3-independent cell death had been blocked by Z-VAD-FMK (pan-caspase inhibitor) or caspase 8 knockdown, demonstrating that RIP3 knockdown switched TNFα-induced necroptosis to caspase-dependent apoptosis. Although both TNF receptor type 1-associated death domain protein (TRADD) and RIP1 have been reported to mediate TNFα-induced apoptosis, the knockdown of TRADD, but not RIP1, suppressed TNFα-induced activation of the caspase pathway and subsequent apoptosis in RIP3 knockdown L929 cells. In addition, TRADD bound and activated caspase 8 during the RIP3-independent apoptosis process, indicating that TRADD initiates RIP3-independent apoptosis by activating the caspase pathway. Collectively, we identified the target and mechanism underlying RIP3-independent apoptosis and elucidated the coordinated roles of RIP3 and TRADD in mediating the programmed cell death of L929 cells following TNFα stimulation.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Apoptosis/genética , Caspasa 8/genética , Caspasa 8/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Ratones , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/efectos de los fármacos , Proteína de Dominio de Muerte Asociada a Receptor de TNF/genética
8.
Nanomaterials (Basel) ; 7(10)2017 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-29036920

RESUMEN

A pattern gel has been fabricated using sodium hyaluronate (HA) and 1,4-butanediol diglycidyl ether (BDDGE) through the micro-molding technique. The cellular behavior of osteoblast cells (MC3T3) in the presence and absence of dimethyloxalylglycine (DMOG) and sodium borate (NaB) in the pattern gel (HA-BDDGE) has been evaluated for its potential application in bone regeneration. The Fourier transform infrared spectroscopy (FTIR), 13C-nuclear magnetic resonance spectroscopy (13C NMR), and thermogravimetric analysis (TGA) results implied the crosslinking reaction between HA and BDDGE. The scanning electron microscopy (SEM) analysis confirmed the formation of pattern on the surface of HA-BDDGE. The gel property of the crosslinked HA-BDDGE has been investigated by swelling study in distilled water at 37 °C. The HA-BDDGE gel releases DMOG in a controlled way for up to seven days in water at 37 °C. The synthesized gel is biocompatible and the bolus drug delivery results indicated that the DMOG containing patterned gel demonstrates a better cell migration ability on the surface than NaB. For local delivery, the pattern gel with 300 µM NaB or 300 µM DMOG induced cell clusters formation, and the gel with 150 µM NaB/DMOG showed high cell proliferation capability only. The vital role of NaB for bone regeneration has been endorsed from the formation of cell clusters in presence of NaB in the media. The in vitro results indicated that the pattern gel showed angiogenic and osteogenic responses with good ALP activity and enhanced HIF-1α, and Runx2 levels in the presence of DMOG and NaB in MC3T3 cells. Hence, the HA-BDDGE gel could be used in bone regeneration application.

9.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(3): 337-341, 2017 Mar.
Artículo en Chino | MEDLINE | ID: mdl-28274312

RESUMEN

Objective To establish a human bladder cancer cell line stably co-expressing human sprouty2 (hSPRY2) and luciferase (Luc) genes simultaneously, and develop its subcutaneous tumor xenograft model in nude mice. Methods The hSPRY2 and Luc gene segments were amplified by PCR, and were cloned into lentiviral vector pCDH and pLVX respectively to produce corresponding lentivirus particles. The J82 human bladder cancer cells were infected with these two kinds of lentivirus particles, and then further screened by puromycin and G418. The expressions of hSPRY2 and Luc genes were detected by bioluminescence, immunofluorescence and Western blot analysis. The screened J82-hSPRY2/Luc cells were injected subcutaneously into BALB/c nude mice, and the growth of tumor was monitored dynamically using in vivo fluorescence imaging system. Results J82-hSPRY2/Luc cell line stably expressing hSPRY2 and Luc genes was established successfully. Bioluminescence, immunofluorescence and Western blot analysis validated the expressions of hSPRY2 and Luc genes. The in vivo fluorescence imaging system showed obvious fluorescence in subcutaneous tumor xenograft in nude mice. Conclusion The J82-hSPRY2/Luc bladder cancer cell line and its subcutaneous tumor xenograft model in nude mice have been established successfully.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Luciferasas/genética , Proteínas de la Membrana/genética , Neoplasias de la Vejiga Urinaria/genética , Animales , Línea Celular Tumoral , Genes Reporteros , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Luciferasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Vejiga Urinaria/metabolismo
10.
Sci Rep ; 6: 38267, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27905569

RESUMEN

The prognosis of gastric cancer remains poor due to clinical drug resistance. Novel drugs are urgently needed. Shikonin (SHK), a natural naphthoquinone, has been reported to trigger cell death and overcome drug resistance in anti-tumour therapy. In this study, we investigated the effectiveness and molecular mechanisms of SHK in treatment with gastric cancer. In vitro, SHK suppresses proliferation and triggers cell death of gastric cancer cells but leads minor damage to gastric epithelial cells. SHK induces the generation of intracellular reactive oxygen species (ROS), depolarizes the mitochondrial membrane potential (MMP) and ultimately triggers mitochondria-mediated apoptosis. We confirmed that SHK induces apoptosis of gastric cancer cells not only in a caspase-dependent manner which releases Cytochrome C and triggers the caspase cascade, but also in a caspase-independent manner which mediates the nuclear translocation of apoptosis-inducing factor and Endonuclease G. Furthermore, we demonstrated that SHK enhanced the chemotherapeutic sensitivity of 5-fluorouracil and oxaliplatin in vitro and in vivo. Taken together, our data show that SHK may be a novel therapeutic agent in the clinical treatment of gastric cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Mitocondrias/metabolismo , Naftoquinonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Línea Celular Tumoral , Fluorouracilo/farmacología , Humanos , Mitocondrias/patología , Compuestos Organoplatinos/farmacología , Piridinas/farmacología , Neoplasias Gástricas/patología
11.
Onco Targets Ther ; 8: 3067-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26604780

RESUMEN

BACKGROUND: Persistent infection with high-risk human papillomavirus (HPV) is a predominant cause of cervical cancer, and HPV58 is the third most common virus detected in the patients with cervical cancer in Asia. E6 and E7 are the viral oncogenes which are constitutively expressed in HPV-associated tumor cells and can be used as target antigens for related immunotherapy. In this study, we modified the HPV58 E6 and E7 oncogenes to eliminate their oncogenic potential and constructed a recombinant DNA vaccine that coexpresses the sig-HPV58 mE6E7-Fc-GPI fusion antigen in addition to granulocyte-macrophage colony-stimulating factor (GM-CSF) and B7.1 as molecular adjuvants (PVAX1-HPV58 mE6E7FcGB) for the treatment of HPV58 (+) cancer. METHODS: PVAX1-HPV58 mE6E7FcGB recombinant DNA vaccine was constructed to express a fusion protein containing a signal peptide, a modified HPV58 mE6E7 gene, and human IgG Fc and glycosylphosphatidylinositol (GPI)-anchoring sequences using the modified DNA vaccine vector PVAX1-IRES-GM/B7.1 that coexpresses GM-CSF, and B7.1. C57BL/6 mice were challenged by HPV58 E6E7-expressing B16-HPV58 E6E7 cells, followed by immunization by PVAX1-HPV58 mE6E7FcGB vaccine on days 7, 14, 21 after tumor challenge. The cellular immune responses in immunized mice were assessed by measuring IFN-γ production in splenocytes upon stimulation by HPV58 E6E7-GST protein and the lysis of B16-HPV58 E6E7 target cells by splenocytes after restimulation with HPV58 E6E7-GST protein. The antitumor efficacy was evaluated by monitoring the growth of the tumor. RESULTS: PVAX1-HPV58 mE6E7FcGB elicited varying levels of IFN-lsgdB58onn T-cell immune responses and lysis of target cell in mice in response to the recombinant antigen HPV58 E6E7-GST. Furthermore, the vaccine also induced antitumor responses in the HPV58 (+) B16-HPV58 E6E7 tumor challenge model as evidenced by delayed tumor development. CONCLUSION: The recombinant DNA vaccine PVAX1-HPV58 mE6E7FcGB efficiently generates cellular immunity and antitumor efficacy in immunized mice. These data provide a basis for the further study of this recombinant vaccine as a potential candidate vaccine.

12.
Mol Immunol ; 67(2 Pt B): 532-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26265114

RESUMEN

AKT and ERK pathways are known to be activated in human rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS), which play crucial roles in the pathogenesis and joint destruction of RA. Sprouty2 (SPRY2) has been known as a tumor suppressor by preventing both ERK and AKT signaling activations. Whether SPRY2 can function as a suppressor in tumor-like inflammatory FLS through negatively regulating AKT and ERK pathways, has not been reported. The purpose of this study was to determine whether SPRY2 might have antiinflammatory effects on RA FLS. The recombinant adenovirus containing SPRY2 complementary DNA (AdSPRY2) was used to deliver SPRY2 and express the protein in RA FLS. Adenoviral vector encoding green fluorescent protein (AdGFP) was used as the control. AdSPRY2 treatment suppressed the production of proinflammatory cytokines and matrix metalloproteinases (MMPs), and the cell proliferation, induced by TNFα in RA FLS. SPRY2 overexpression reduced AKT and ERK phosphorylation in TNFα-stimulated FLS, through mediating or interfering with the activity of PTEN or Raf respectively. These results suggest that using SPRY2 to block the AKT and ERK pathways suppresses the inflammatory responses of RA FLS, and the development of an immunoregulatory strategy based on SPRY2 may therefore have therapeutic potential in the treatment of RA.


Asunto(s)
Artritis Reumatoide/patología , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Membrana Sinovial/patología , Quinasas raf/metabolismo , Adenoviridae/metabolismo , Artritis Reumatoide/genética , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Mediadores de Inflamación/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de la Membrana/genética , Fosforilación/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
13.
Biochem Biophys Res Commun ; 465(2): 239-44, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26253468

RESUMEN

To further enhance the antitumor efficacy of DNA vaccine, we proposed a synergistic strategy that targeted tumor cells and angiogenesis simultaneously. In this study, a Semliki Forest Virus (SFV) replicon DNA vaccine expressing 1-4 domains of murine VEGFR2 and IL12 was constructed, and was named pSVK-VEGFR2-GFc-IL12 (CAVE). The expression of VEGFR2 antigen and IL12 adjuvant molecule in 293T cells in vitro were verified by western blot and enzyme-linked immune sorbent assay (ELISA). Then CAVE was co-immunized with CAVA, a SFV replicon DNA vaccine targeting survivin and ß-hCG antigens constructed previously. The antitumor efficacy of our combined replicon vaccines was evaluated in mice model and the possible mechanism was further investigated. The combined vaccines could elicit efficient humoral and cellular immune responses against survivin, ß-hCG and VEGFR2 simultaneously. Compared with CAVE or CAVA vaccine alone, the combined vaccines inhibited the tumor growth and improved the survival rate in B16 melanoma mice model more effectively. Furthermore, the intratumoral microvessel density was lowest in combined vaccines group than CAVE or CAVA alone group. Therefore, this synergistic strategy of DNA vaccines for tumor treatment results in an increased antitumor efficacy, and may be more suitable for translation to future research and clinic.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Melanoma Experimental/terapia , Neovascularización Patológica/prevención & control , Neoplasias Cutáneas/terapia , Vacunas de ADN/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Gonadotropina Coriónica Humana de Subunidad beta/antagonistas & inhibidores , Gonadotropina Coriónica Humana de Subunidad beta/genética , Gonadotropina Coriónica Humana de Subunidad beta/inmunología , Femenino , Expresión Génica , Células HEK293 , Humanos , Inmunización , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/inmunología , Interleucina-12/antagonistas & inhibidores , Interleucina-12/genética , Interleucina-12/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Plásmidos/química , Plásmidos/metabolismo , Replicón , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Survivin , Resultado del Tratamiento , Vacunas Combinadas , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
14.
Appl Microbiol Biotechnol ; 99(16): 6727-35, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25935347

RESUMEN

AKT and ERK pathways have been implicated as therapeutic targets for human rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) inhibition, and thus RA treatment. Sprouty2 (SPRY2) has been known as a tumor suppressor by blocking both ERK and AKT signaling cascades. Whether SPRY2 can function as a suppressor of tumor-like inflammatory FLS and RA through negatively regulating AKT and ERK activation has not been reported. The purpose of this study was to determine whether SPRY2 might have antiarthritic effects in experimental animal model of RA. We first determined that expression of SPRY2 mRNA was decreased in FLS from patients with RA compared with patients with osteoarthritis (OA). Further studies demonstrated that intraarticular gene transfer with AdSPRY2, the recombinant adenovirus containing SPRY2 complementary DNA, resulted in a significant suppression of rat adjuvant-induced arthritis (AIA) compared with the control AdGFP, the adenoviral vector encoding green fluorescent protein, as reflected in both clinical and histological observations. AdSPRY2 suppressed the production of proinflammatory cytokines and matrix metalloproteinases (MMPs), and the activation of ERK and AKT signals in AIA ankle joints. These results suggest that using SPRY2 to block the AKT and ERK pathways effectively reduces the inflammatory responses and arthritic progression in AIA. Thus, the development of an immunoregulatory strategy based on SPRY2 may therefore have therapeutic potential in the treatment of RA.


Asunto(s)
Artritis Reumatoide/patología , Artritis Reumatoide/terapia , Terapia Genética/métodos , Proteínas del Tejido Nervioso/metabolismo , Adenoviridae/genética , Animales , Artritis Reumatoide/inducido químicamente , Citocinas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Vectores Genéticos , Humanos , Sistema de Señalización de MAP Quinasas , Metaloproteinasas de la Matriz/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Ratas , Resultado del Tratamiento
15.
Appl Microbiol Biotechnol ; 99(15): 6327-37, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25707864

RESUMEN

The complexity of rheumatoid arthritis (RA) pathogenesis makes combined blockade of key pathogenic factors an attractive therapeutic strategy. We have previously reported a novel recombinant adeno-associated virus (AAV) vector, AAV.TFCF, which mediates separate coexpression of TNFα antagonist TNFR-Fc and T cell antagonist CTLA4-FasL both in vitro and in vivo (the injected joints). The purpose of this study was to examine the efficacy of TNFR-Fc/CTLA4-FasL combination therapy mediated by AAV.TFCF in experimental model of RA. Adjuvant-induced arthritis (AIA) was induced in Lewis rats, and the recombinant AAV.TFCF was injected into rat ankle joints. AAV vector encoding CTLA4-FasL (AAV.CTFA) or TNFR-Fc (AAV.TRFC) was used as the monotherapy control, and an AAV vector mediating the expression of enhanced green fluorescent protein (AAV.EGFP) was used as the negative control. The combination treatment mediated by AAV.TFCF demonstrated a more effective suppression of AIA compared with those monotherapy controls, as reflected in the clinical and histological observations. The synergistic anti-inflammatory effect of TNFR-Fc combining with CTLA4-FasL was proved to be associated with the greater reductions of inflammatory CD4+ T cell infiltration and proinflammatory cytokine TNFα level in the arthritic joints. In addition, the combination therapy was found to be able to increase the frequency of CD4 + CD25 + FoxP3+ regulatory T cell population in rat draining lymph nodes and suppress splenic inflammatory responses. These results suggest that combination treatment with TNFR-Fc and CTLA4-FasL may achieve superior efficacy in suppressing RA, and using this novel recombinant AAV.TFCF to obtain the combined counteraction of both pathogenic T cells and the key proinflammatory cytokine TNFα may provide a more effective and desirable strategy for treatment of RA.


Asunto(s)
Artritis/terapia , Antígeno CTLA-4/uso terapéutico , Dependovirus/genética , Portadores de Fármacos , Etanercept/uso terapéutico , Factores Inmunológicos/uso terapéutico , Inmunoterapia/métodos , Animales , Artritis/patología , Antígeno CTLA-4/genética , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Factores Inmunológicos/genética , Ratas Endogámicas Lew , Transducción Genética , Resultado del Tratamiento
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 30(8): 819-23, 2014 Aug.
Artículo en Chino | MEDLINE | ID: mdl-25108434

RESUMEN

OBJECTIVE: To investigate the effect of p53-regulated apoptosis-inducing protein 1 (p53AIP1) gene on the proliferation, cell cycle, apoptosis, invasion and migration of PC-3M human prostate cancer cells in vitro. METHODS: The eukaryotic expression vector pDC316-p53AIP1 was constructed and confirmed by double enzyme digestion and PCR analysis. Then it was transfected into PC-3M human prostate cancer cells by Lipofectamine (TM) 2000. The expression of p53AIP1 protein was detected by Western blotting. The proliferation of PC-3M cells was tested by CCK-8 assay; the cell cycle and apoptosis rate were analyzed by flow cytometry combined with annexin V-FITC/PI staining; the effect of p53AIP1 on the cell invasion and migration was detected by Transwell(TM) assay. RESULTS The eukaryotic expression vector pDC316-p53AIP1 was constructed successfully. After transfected into PC-3M cells, Western blotting demonstrated that the protein p53AIP1 was effectively expressed. CCK-8 assay revealed that the proliferation of PC-3M cells was significantly inhibited by p53AIP1 (P<0.05). Flow cytometry indicated that the cells were arrested at S/G2-M phase (P<0.05) and cell apoptosis was evidently promoted (P<0.05). Transwell(TM) chamber experiments showed that p53AIP1 decreased the abilities of both invasion and migration of the cells (P<0.05). CONCLUSION: The p53AIP1 inhibits the proliferation of PC-3M cells, arrests cell cycle at S/G2-M phase, decreases the abilities of invasion and migration and promotes cell apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Movimiento Celular/genética , Citometría de Flujo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transfección
17.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 30(5): 476-9, 484, 2014 May.
Artículo en Chino | MEDLINE | ID: mdl-24796741

RESUMEN

OBJECTIVE: To construct a prokaryotic expression plasmid for CT40L, express the target protein in E. coli, purity the CT40L fusion protein and verify its antigenicity. METHODS: Gene sequences of Coxsackie and adenovirus receptor (CAR), bacteriophage T4 fibritin and mouse CD40L were found out in GenBank. Then functional domains of three molecules were linked to form a fusion sequence which was then optimized for prokaryotic expression. The optimized sequence was cloned into prokaryotic expression vector pET42a(+) to construct the recombinant expression vector pET42a-CT40L. The recombinant vector was transformed into BL21 (DE3) and the fusion protein CT40L/GST was induced by IPTG. The fusion protein was then subjected to purification using GST affinity chromatography and to identification of the immune activity using Western blotting and ELISA. RESULTS: The recombinant expression vector was verified correct by double digestion with Nco I and EcoR I. After IPTG induction, SDS-PAGE showed that the relative molecular mass of the fusion protein was about 78 kDa and that the purity of the purified protein reached 90%. Western blotting and ELISA demonstrated that the purified fusion protein had a valid antigenicity. CONCLUSION: The prokaryotic expression plasmid pET42a-Ct40L was successfully constructed and expressed in E. coli, and the purified fusion protein was proved to have a good antigenicity.


Asunto(s)
Ligando de CD40/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Células Dendríticas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Virales/metabolismo , Adenoviridae/genética , Adenoviridae/inmunología , Adenoviridae/fisiología , Secuencia de Aminoácidos , Animales , Western Blotting , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Ligando de CD40/genética , Ligando de CD40/inmunología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Ratones , Datos de Secuencia Molecular , Unión Proteica/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 30(5): 485-8, 2014 May.
Artículo en Chino | MEDLINE | ID: mdl-24796743

RESUMEN

OBJECTIVE: To construct a recombinant expression vector pDC315/hSPRY2 carrying human SPRY2 (hSPRY2) gene, and transfect the recombinant plasmid into human embryonic kidney HEK293T cells to express the target protein, and thus explore preliminarily the effect of hSPRY2 on the proliferation and survival of HEK293T cells. METHODS: The recombinant adenoviral vector expressing hSPRY2 was constructed and transfected into HEK293T cells in vitro. The expression of hSPRY2 target protein in the transfected cells was identified by Western blot analysis. The effect of hSPRY2 on the proliferation and survival of HEK293T cells in a serum-free condition was tested using CCK-8 assay. RESULTS: The recombinant expression vector pDC315/hSPRY2 was constructed, and the target protein hSPRY2 was expressed in the transfected HEK293T cells. The proliferation and survival rates of HEK293T cells transfected with pDC315/hSPRY2 were significantly higher than those transfected with pDC315/EGFP (P<0.05). CONCLUSION: The recombinant expression vector for hSPRY2 was successfully constructed and its expression was demonstrated in the transfected HEK293T cells. Over-expression of SPRY2 promoted the proliferation and survival of HEK293T cells.


Asunto(s)
Proliferación Celular , Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Western Blotting , Supervivencia Celular/genética , Clonación Molecular/métodos , Citometría de Flujo , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
19.
PLoS One ; 9(3): e90551, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24608380

RESUMEN

A DNA-based replicon vaccine derived from Semliki Forest virus, PSVK-shFcG-GM/B7.1 (Fig. 1a) was designed for tumor immunotherapy as previously constructed. The expression of the fusion tumor antigen (survivin and hCGß-CTP37) and adjuvant molecular protein (Granulocyte-Macrophage Colony-Stimulating Factor/ GM-CSF/B7.1) genes was confirmed by Immunofluorescence assay in vitro, and immunohistochemistry assay in vivo. In this paper, the immunological effect of this vaccine was determined using immunological assays as well as animal models. The results showed that this DNA vaccine induced both humoral and cellular immune responses in C57BL/6 mice after immunization, as evaluated by the ratio of CD4+/CD8+ cells and the release of IFN-γ. Furthermore, the vaccination of C57BL/6 mice with PSVK-shFcG-GM/B7.1 significantly delayed the in vivo growth of tumors in animal models (survivin+ and hCGß+ murine melanoma, B16) when compared to vaccination with the empty vector or the other control constructs (Fig. 1b). These data indicate that this type of replicative DNA vaccine could be developed as a promising approach for tumor immunotherapy. Meanwhile, these results provide a basis for further study in vaccine pharmacodynamics and pharmacology, and lay a solid foundation for clinical application.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Replicón/inmunología , Virus de los Bosques Semliki/inmunología , Vacunas de ADN/inmunología , Animales , Línea Celular Tumoral , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Ratones , Ratones Endogámicos C57BL
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 30(3): 225-8, 2014 Mar.
Artículo en Chino | MEDLINE | ID: mdl-24606734

RESUMEN

OBJECTIVE: To construct a recombinant adeno-associated virus vector carrying the fusion product of extracellular domain of human tumor necrosis factor receptor (hTNFR) with human IgG Fc fragment, and detect the expression and biological activity of hTNFR-Fc. METHODS: The recombinant hTNFR-Fc fusion gene was cloned into adeno-associated virus (AAV) vector, which was used to transfect human embryo kidney cells (HEK293T) in vitro. The expression of the target protein hTNFR-Fc in the supernatants of the transfected HEK293T cells was identified by Western blot analysis. TNF-α inhibition assay in vitro was performed using TNF-α-sensitive mouse fibrosarcoma L929 cells to determine the neutralizing capacity of hTNFR-Fc against human or rat TNF-α. The binding activity of hTNFR-Fc to human or rat TNF-α was identified by ELISA. RESULTS: The recombinant plasmid pAAV/hTNFR-Fc was constructed and the target protein of hTNFR-Fc was expressed in the supernatants of the transfected HEK293T cells. It could effectively bind to human TNF-α and neutralize its cytotoxicity to L929 cells. The hTNFR-Fc protein also exerted the binding and neutralization activities to rat TNF-α to some extent. CONCLUSION: The study constructed hTNFR-Fc fusion gene and verified its secretory expression in AAV vector, as well as identified its biological activities, which lay the foundations for future viral package and animal experiments.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/genética , Receptores del Factor de Necrosis Tumoral/genética , Proteínas Recombinantes de Fusión/biosíntesis , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Dependovirus/genética , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Ratas , Proteínas Recombinantes de Fusión/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA