Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 591(7851): 645-651, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33589820

RESUMEN

Regulatory T (Treg) cells, although vital for immune homeostasis, also represent a major barrier to anti-cancer immunity, as the tumour microenvironment (TME) promotes the recruitment, differentiation and activity of these cells1,2. Tumour cells show deregulated metabolism, leading to a metabolite-depleted, hypoxic and acidic TME3, which places infiltrating effector T cells in competition with the tumour for metabolites and impairs their function4-6. At the same time, Treg cells maintain a strong suppression of effector T cells within the TME7,8. As previous studies suggested that Treg cells possess a distinct metabolic profile from effector T cells9-11, we hypothesized that the altered metabolic landscape of the TME and increased activity of intratumoral Treg cells are linked. Here we show that Treg cells display broad heterogeneity in their metabolism of glucose within normal and transformed tissues, and can engage an alternative metabolic pathway to maintain suppressive function and proliferation. Glucose uptake correlates with poorer suppressive function and long-term instability, and high-glucose conditions impair the function and stability of Treg cells in vitro. Treg cells instead upregulate pathways involved in the metabolism of the glycolytic by-product lactic acid. Treg cells withstand high-lactate conditions, and treatment with lactate prevents the destabilizing effects of high-glucose conditions, generating intermediates necessary for proliferation. Deletion of MCT1-a lactate transporter-in Treg cells reveals that lactate uptake is dispensable for the function of peripheral Treg cells but required intratumorally, resulting in slowed tumour growth and an increased response to immunotherapy. Thus, Treg cells are metabolically flexible: they can use 'alternative' metabolites in the TME to maintain their suppressive identity. Further, our results suggest that tumours avoid destruction by not only depriving effector T cells of nutrients, but also metabolically supporting regulatory populations.


Asunto(s)
Ácido Láctico/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Glucosa/metabolismo , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Factores Supresores Inmunológicos/inmunología , Factores Supresores Inmunológicos/metabolismo , Linfocitos T Reguladores/inmunología
2.
Mol Biol Rep ; 49(2): 1213-1222, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34773179

RESUMEN

BACKGROUND: Quercetin is a flavonol that modifies many cellular processes. Monoclonal nonspecific suppressor factor ß is a member of the ubiquitin-like family of proteins that are involved in various biological processes. It has been demonstrated that quercetin regulates the effect of MNSFß on tumor necrosis factor-α secretion in lipopolysaccharide (LPS)-stimulated macrophages. This study found that quercetin and the heat shock protein HSC70 coregulate the action of MNSFß. METHODS AND RESULTS: Quercetin dose-dependently suppressed the LPS/interferon γ-induced nitric oxide production without cytotoxicity in the macrophage-like cell line Raw264.7. SiRNA knockdown experiments showed that quercetin inhibited the MNSFß and HSC70 siRNA-mediated enhancement of TNFα and the production of RANTES, a member of C-C chemokine superfamily, in LPS-stimulated Raw264.7 cells. Western blot analysis showed that quercetin and HSC70 regulated ERK1/2 activation and LPS-stimulated IκBα degradation by affecting the complex formation of MNSFß and the proapoptotic protein Bcl-G. Moreover, MNSFß is implicated in TLR4/MyD88 signaling but not in TLR3 signaling. CONCLUSIONS: HSC70 is an important chaperone that facilitates the stabilization of MNSFß. Quercetin may negatively control the function of MNSFß by regulating the action of the molecular chaperone HSC70. MNSFß mediates TLR4/Myd88 signaling but not TLR3 signaling.


Asunto(s)
Proteínas del Choque Térmico HSC70/metabolismo , Quercetina/farmacología , Factores Supresores Inmunológicos/metabolismo , Animales , Línea Celular , Flavonoides/farmacología , Proteínas del Choque Térmico HSC70/efectos de los fármacos , Interferón gamma/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Óxido Nítrico/metabolismo , Quercetina/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/metabolismo
3.
Gynecol Obstet Invest ; 86(1-2): 27-39, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33326956

RESUMEN

AIMS: The invasion of extravillous trophoblast (EVT) cells into maternal decidua is essential for the establishment and maintenance of pregnancy. Derangement of EVT cell invasion might cause pregnancy complications including recurrent miscarriage (RM). We previously reported that deficiency of monoclonal nonspecific suppressor factor beta (MNSFß) led to the early pregnancy failure in mice and the decidual MNSFß expression level in RM patients was significantly decreased, but the underlying molecular mechanism of the role that MNSFß played at the maternal-fetal interface remains unclear. Thus, in the present study, we determined effects of downregulated MNSFß expression on human EVT cell activities. METHODS: The MNSFß expression in first-trimester human decidual and placental villus tissues was detected, respectively, by immunofluorescence or immunohistochemical analyses. The MNSFß expression level in the immortalized first-trimester human EVT cell line HTR8/SVneo was downregulated by transfecting the small interfering RNA against MNSFß and upregulated by transfecting the recombinant pDsRed-MNSFß plasmids. The proliferation, migration, invasion, and apoptosis activities of HTR8/SVneo cells were, respectively, determined by cytometry assay, scratch test, transwell assay, and FITC/PI staining. The expression levels of P53, RhoA, Bcl-2, Bax, and MMP-9 in HTR8/SVneo cells, as well as the expression levels of MNSFß and RhoA in placental villi of RM patients and physically normal pregnant women (NP), were examined by Western blot analysis. RESULTS: MNSFß protein signals were observed in first-trimester human villus and extravillous trophoblast cells. The downregulated MNSFß expression significantly attenuated the proliferation, migration, and invasion abilities of HTR8/SVneo cells, accompanied with the obviously decreased expression levels of P53, RhoA, Bcl-2, Bax, and MMP-9, whereas the upregulated MNSFß expression in HTR8/SVneo cells represented the inverse effects. Furthermore, expression levels of MNSFß and RhoA in first-trimester human placental villus tissues of RM patients were significantly decreased compared to that of NP women. CONCLUSION: These data suggested that MNSFß promotes proliferation and migration of human EVT cells, probably via the P53 signaling pathway, and the deficiency of MNSFß in placental villi might lead to early pregnancy loss by reducing proliferation and invasion activities of EVTs.


Asunto(s)
Apoptosis , Movimiento Celular , Proliferación Celular , Factores Supresores Inmunológicos/metabolismo , Trofoblastos/metabolismo , Aborto Habitual , Animales , Línea Celular , Vellosidades Coriónicas , Regulación hacia Abajo , Femenino , Genes bcl-2 , Genes p53 , Humanos , Metaloproteinasa 9 de la Matriz , Ratones , Embarazo , Primer Trimestre del Embarazo , ARN Interferente Pequeño , Transducción de Señal , Proteína X Asociada a bcl-2 , Proteína de Unión al GTP rhoA/metabolismo
4.
Breast Cancer Res Treat ; 182(3): 591-600, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32529408

RESUMEN

PURPOSE: This study evaluates the oncogenic role of PIBF1 in triple-negative breast cancer (TNBC). TNBC is considered to have a poorer prognosis than other types of breast cancer and is associated with high risk of recurrence and distant metastasis. Currently, there are no effective therapies for the TNBC patients with distant metastasis due to the lack of targeted therapeutic options. METHODS: The effects of PIBF1 knockdown on the cell viability and motility of TNBC cell lines were investigated. Effects of PIBF1 overexpression on tumorigenicity and cell motility were confirmed using Ba/F3 cell line and xenograft study on BALB/c nude mice. RESULTS: In TNBC cell lines that highly express PIBF1, knockdown of PIBF1 induces apoptosis and suppresses cell viability and motility with activation of the ATR/CHK1 signaling pathway. Moreover, the oncogenic function of PIBF1 was confirmed using the Ba/F3 cell line. CONCLUSION: For the first time, these findings clarify the role of PIBF1 in regulating ATR/CHK1 signaling pathway and inhibiting the proliferation and migration of TNBC cell lines. These results demonstrate the oncogenic roles of PIBF1 and provide new insights into the function and the molecular mechanism of PIBF1 in malignant TNBC.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Proteínas Gestacionales/metabolismo , Factores Supresores Inmunológicos/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Apoptosis/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Gestacionales/biosíntesis , Proteínas Gestacionales/genética , Transducción de Señal , Factores Supresores Inmunológicos/biosíntesis , Factores Supresores Inmunológicos/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas
5.
Mol Cell Biochem ; 456(1-2): 29-39, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30710197

RESUMEN

Monoclonal nonspecific suppressor factor ß (MNSFß) is a ubiquitously expressed ubiquitin-like protein known to be involved in various biological functions. Previous studies have demonstrated that MNSFß covalently modify its target proteins including Bcl-G, a proapoptotic protein. In this study, we purified a 65 kDa MNSFß adduct from mouse liver lysates by sequential chromatography on DEAE and glutathione S-transferase (GST)-fusioned MNSFß immobilized on glutathione-Sepharose beads in the presence of ATP. MALDI-TOF mass spectrometry fingerprinting revealed that this MNSFß adduct consists of an 8.5 kDa MNSFß and heat shock protein 60 (HSP60), a mitochondrial protein involved in protein folding. Fingerprinting analysis of the MNSFß adduct demonstrates that MNSFß conjugates to HSP60 with a linkage between the C-terminal Gly74 and Lys481. HSP60 siRNA neutralized the inhibition of apoptosis by MNSFß siRNA in LPS/IFNγ-stimulated Raw264.7, a murine macrophage cell line. HSP60 siRNA also down-regulated the enhancement of TNFα production by MNSFß siRNA in LPS-stimulated Raw264.7 cells. Here, we firstly report that MNSFß activity is negatively regulated by molecular chaperone.


Asunto(s)
Chaperonina 60/metabolismo , Macrófagos/metabolismo , Proteínas Mitocondriales/metabolismo , Pliegue de Proteína , Factores Supresores Inmunológicos/metabolismo , Animales , Lipopolisacáridos/toxicidad , Ratones , Células RAW 264.7
6.
J Biol Chem ; 290(49): 29290-300, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26429916

RESUMEN

Precise modulation of histone gene transcription is critical for cell cycle progression. As a direct substrate of Cyclin E/CDK2, nuclear protein ataxia-telangiectasia (NPAT) is a crucial factor in regulating histone transcription and cell cycle progression. Here we identified that Cpn10/HSPE, a 10-kDa heat shock protein, is a novel interacting partner of NPAT. A pool of Cpn10 is colocalized with NPAT foci during G1 and S phases in nuclei. Gain- and loss-of-function experiments unraveled an essential role of Cpn10 in histone transcription. A conserved DLFD motif within Cpn10 was critical for targeting NPAT and modulating histone transcription. More importantly, knockdown of Cpn10 disrupted the focus formation of both NPAT and FADD-like interleukin-1ß-converting enzyme-associated huge protein without affecting Coilin-positive Cajal bodies. Finally, Cpn10 is important for S phase progression and cell proliferation. Taken together, our finding revealed a novel role of Cpn10 in the spatial regulation of NPAT signaling and disclosed a previously unappreciated link between the heat shock protein and histone transcription regulation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Chaperonina 10/metabolismo , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Gestacionales/metabolismo , Factores Supresores Inmunológicos/metabolismo , Secuencias de Aminoácidos , Ciclo Celular , Núcleo Celular/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Células HeLa , Humanos , Microscopía Fluorescente , Interferencia de ARN , Transducción de Señal , Transcripción Genética , Técnicas del Sistema de Dos Híbridos
7.
Mol Cell Biochem ; 421(1-2): 149-56, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27581120

RESUMEN

MNSFß, a ubiquitin-like protein, covalently binds to various target proteins including proapoptotic Bcl-G. During the course of isolation of MNSFß-conjugating enzyme(s), we identified a novel target protein for MNSFß. MALDI-TOF MS fingerprinting revealed that the MNSFß-interacting protein is HSPA8 (heat shock 70-kDa protein 8). We observed that MNSFß noncovalently binds to HSPA8 in the presence of ATP in vitro. Double knockdown of MNSFß and HSPA8 strongly inhibited RANKL-induced osteoclastogenesis from Raw264.7 macrophage-like cells. The same treatment inhibited RANKL-induced ERK1/2 and p38 phosphorylation and TNFα production, suggesting that the association of MNSFß with HSPA8 may promote RANKL-induced osteoclastogenesis. This is the first report that MNSFß binds to a protein substrate via the noncovalent association and exerts biological effects.


Asunto(s)
Proteínas del Choque Térmico HSC70/metabolismo , Sistema de Señalización de MAP Quinasas , Osteoclastos/metabolismo , Factores Supresores Inmunológicos/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Proteínas del Choque Térmico HSC70/química , Ratones , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Unión Proteica , Ligando RANK/metabolismo , Factores Supresores Inmunológicos/química
8.
Nat Rev Immunol ; 5(8): 641-54, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16056256

RESUMEN

L-Arginine is an essential amino acid for birds and young mammals, and it is a conditionally essential amino acid for adult mammals, as it is important in situations in which requirements exceed production, such as pregnancy. Recent findings indicate that increased metabolism of L-arginine by myeloid cells can result in the impairment of lymphocyte responses to antigen during immune responses and tumour growth. Two enzymes that compete for L-arginine as a substrate - arginase and nitric-oxide synthase - are crucial components of this lymphocyte-suppression pathway, and the metabolic products of these enzymes are important moderators of T-cell function. This Review article focuses on the relevance of L-arginine metabolism by myeloid cells for immunity under physiological and pathological conditions.


Asunto(s)
Arginina/metabolismo , Células Mieloides/enzimología , Linfocitos T/inmunología , Animales , Arginasa/metabolismo , Humanos , Inmunidad Celular , Óxido Nítrico Sintasa/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores Supresores Inmunológicos/metabolismo
9.
Biochim Biophys Acta ; 1838(1 Pt B): 148-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23954806

RESUMEN

Receptor-ligand binding is an essential interaction for biological function. Oxidative stress can modify receptors and/or membrane lipid dynamics, thus altering cell physiological functions. The aim of this study is to analyze how oxidative stress may alter receptor-ligand binding and lipid domain distribution in the case of progesterone-induced blocking factor/progesterone-induced blocking factor-receptor. For membrane fluidity regionalization analysis of MEC-1 lymphocytes, two-photon microscopy was used in individual living cells. Lymphocytes were also double stained with AlexaFluor647/progesterone-induced blocking factor and Laurdan to evaluate -induced blocking factor/progesterone-induced blocking factor-receptor distribution in the different membrane domains, under oxidative stress. A new procedure has been developed which quantitatively analyzes the regionalization of a membrane receptor among the lipid domains of different fluidity in the plasma membrane. We have been able to establish a new tool which detects and evaluates lipid raft clustering from two-photon microscopy images of individual living cells. We show that binding of progesterone-induced blocking factor to progesterone-induced blocking factor-receptor causes a rigidification of plasma membrane which is related to an increase of lipid raft clustering. However, this clustering is inhibited under oxidative stress conditions. In conclusion, oxidative stress decreases membrane fluidity, impairs receptor-ligand binding and reduces lipid raft clustering.


Asunto(s)
Linfocitos B/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Microdominios de Membrana/efectos de los fármacos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Proteínas Gestacionales/química , Receptores de Citocinas/química , Análisis de la Célula Individual/métodos , Factores Supresores Inmunológicos/química , 2-Naftilamina/análogos & derivados , 2-Naftilamina/química , Linfocitos B/citología , Linfocitos B/metabolismo , Carbocianinas/química , Línea Celular Tumoral , Colorantes Fluorescentes/química , Humanos , Lauratos/química , Fluidez de la Membrana/efectos de los fármacos , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Estrés Oxidativo , Proteínas Gestacionales/metabolismo , Unión Proteica , Receptores de Citocinas/metabolismo , Coloración y Etiquetado/métodos , Factores Supresores Inmunológicos/metabolismo
10.
Biochem Biophys Res Commun ; 464(4): 1096-1100, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26192119

RESUMEN

MNSFß is a ubiquitously expressed member of the ubiquitin-like family that has been involved in various biological functions. Previous studies have demonstrated that MNSFß covalently binds to various target proteins including Bcl-G, a proapoptotic protein. In this study, we purified a 115 kDa MNSFß adduct from murine liver lysates by sequential chromatography on DEAE and anti-MNSFß IgG-conjugated Sepharose in the presence of ATP. MALDI-TOF MS fingerprinting revealed that this MNSFß adduct consists of an 8.5 kDa MNSFß and 10-formyltetrahydrofolate dehydrogenase (FDH), an abundant enzyme of folate metabolism. Interestingly, MNSFß preferably binds to cytosolic but not mitochondrial FDH. Fingerprinting analysis of the MNSFß adduct demonstrate that MNSFß conjugates to cytosolic FDH with a linkage between the C-terminal Gly74 and Lys72. The 115 kDa MNSFß/FDH complex was not expressed in any of the tissues examined, indicating that this adduct formation is not ubiquitous. We found that MNSFß/FDH complex formation was induced by dexamethasone in thymocytes. Double knockdown of MNSFß and FDH strongly reduced dexamethasone-induced apoptosis. Collectively, MNSFß/FDH complex formation may positively regulate apoptosis in thymocytes.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Factores Supresores Inmunológicos/metabolismo , Timocitos/citología , Timocitos/metabolismo , Ubiquitinas/metabolismo , Animales , Apoptosis/fisiología , Células Cultivadas , Citosol/enzimología , Ratones , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/química , Unión Proteica , Factores Supresores Inmunológicos/química , Ubiquitinas/química
11.
Immunol Invest ; 44(1): 1-12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25180634

RESUMEN

Monoclonal non-specific suppressor factor ß (MNSFß) is a ubiquitously expressed member of the ubiquitin-like family that is involved in various biological functions. Previous studies have demonstrated that MNSFß covalently binds to intracellular pro-apoptotic protein Bcl-G and regulates apoptosis in macrophages. In this study, we demonstrate that MNSFß negatively regulates T cell function. In murine T-helper type 2 clone, D10.G4.1 (D10) cells transfected with MNSFß cDNA, CD3/CD28-induced ERK1/2 phosphorylation leading to IL-4 production was significantly inhibited. The formation of MNSFß-Bcl-G complex was induced by the CD3/CD28 stimulation. Co-transfection with MNSFß and Bcl-G greatly enhanced CD3/CD28-induced apoptosis in D10 cells. Similarly, co-over-expression of MNSFß and Bcl-G caused a marked enhancement of apoptosis in purified splenic T cells. Interestingly, this MNSFß adduct was also induced in T cells derived from DO11.10 mice stimulated with antigen. Collectively, CD3/CD28-inducible MNSFß-Bcl-G complex may be involved in the regulation of T cell function and survival.


Asunto(s)
Antígenos CD28/genética , Complejo CD3/genética , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Factores Supresores Inmunológicos/genética , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Antígenos CD28/metabolismo , Complejo CD3/metabolismo , Línea Celular , Proliferación Celular , Supervivencia Celular , Interleucina-4/biosíntesis , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Cultivo Primario de Células , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Factores Supresores Inmunológicos/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Transfección
12.
Cell Mol Neurobiol ; 34(4): 479-89, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24474429

RESUMEN

Glioblastoma multiforme (GBM) is the most common and malignant tumor in the central nervous system. One of the contemporary hypotheses postulates that its pathogenesis is associated with the cancer stem cells (CSCs) which originate from mutations in the normal neural stem cells residing in their specific "niches." Simultaneously with its aggressive development the tumor suppresses the local immune system by different secreted and/or cell expressed factors. Progesterone-induced blocking factor (PIBF) is an immunomodulatory protein with known role in the regulation of the immune response in the reproductive system. Expression of PIBF has been described in some tumors as one of the factors suppressing the anti-tumor immunity. The aim of the present study was to check for the expression of PIBF from cells isolated from six GBMs. To characterize the cultured cells and to study the PIBF expression confocal microscopy, flow cytometry, ELISA, and real-time PCR were used. The results obtained showed expression of markers typical for cancer CSCs and secretion of interleukin 6 by the GBM-derived cultured cells. The results convincingly prove that PIBF is intracellularly expressed by the cultured cells from the all six GBM samples, and this fact is confirmed by three different methods-flow cytometry, confocal microscopy, and real-time PCR. This paper reports for the first time the expression of PIBF by GBM-derived cells cultured in vitro and reveals a new aspect of the immunosuppressive mechanism used by GBM in escaping the immune control.


Asunto(s)
Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Gestacionales/metabolismo , Progesterona/metabolismo , Factores Supresores Inmunológicos/metabolismo , Separación Celular , Glioblastoma/patología , Humanos , Inmunohistoquímica/métodos , Células Madre Neoplásicas/citología , ARN Mensajero/metabolismo , Células Tumorales Cultivadas
13.
Cell Mol Life Sci ; 70(23): 4617-30, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23807209

RESUMEN

Invasiveness is a common feature of trophoblast and tumors; however, while tumor invasion is uncontrolled, trophoblast invasion is strictly regulated. Both trophoblast and tumor cells express high levels of the immunomodulatory progesterone-induced blocking factor (PIBF), therefore, we aimed to test the possibility that PIBF might be involved in invasion. To this aim, we used PIBF-silenced or PIBF-treated trophoblast (HTR8/Svneo, and primary trophoblast) and tumor (HT-1080, A549, HCT116, PC3) cell lines. Silencing of PIBF increased invasiveness as well as MMP-2,-9 secretion of HTR8/SVneo, and decreased those of HT-1080 cells. PIBF induced immediate STAT6 activation in both cell lines. Silencing of IL-4Rα abrogated all the above effects of PIBF, suggesting that invasion-related signaling by PIBF is initiated through the IL-4Rα/PIBF-receptor complex. In HTR-8/SVneo, PIBF induced fast, but transient Akt and ERK phosphorylation, whereas in tumor cells, PIBF triggered sustained Akt, ERK, and late STAT3 activation. The late signaling events might be due to indirect action of PIBF. PIBF induced the expression of EGF and HB-EGF in HT-1080 cells. The STAT3-activating effect of PIBF was reduced in HB-EGF-deficient HT-1080 cells, suggesting that PIBF-induced HB-EGF contributes to late STAT3 activation. PIBF binds to the promoters of IL-6, EGF, and HB-EGF; however, the protein profile of the protein/DNA complex is different in the two cell lines. We conclude that in tumor cells, PIBF induces proteins, which activate invasion signaling, while-based on our previous data-PIBF might control trophoblast invasion by suppressing proinvasive genes.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas Gestacionales/metabolismo , Factores Supresores Inmunológicos/metabolismo , Trofoblastos/metabolismo , Animales , Animales Modificados Genéticamente , Western Blotting , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Trasplante de Células/métodos , Células Cultivadas , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HCT116 , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Microscopía Confocal , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Gestacionales/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Transducción de Señal/genética , Factores Supresores Inmunológicos/genética , Trasplante Heterólogo , Trofoblastos/citología , Trofoblastos/trasplante , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
14.
Nat Commun ; 15(1): 1487, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38374152

RESUMEN

Proper placental development in early pregnancy ensures a positive outcome later on. The developmental relationship between the placenta and embryonic organs, such as the heart, is crucial for a normal pregnancy. However, the mechanism through which the placenta influences the development of embryonic organs remains unclear. Trophoblasts fuse to form multinucleated syncytiotrophoblasts (SynT), which primarily make up the placental materno-fetal interface. We discovered that endogenous progesterone immunomodulatory binding factor 1 (PIBF1) is vital for trophoblast differentiation and fusion into SynT in humans and mice. PIBF1 facilitates communication between SynT and adjacent vascular cells, promoting vascular network development in the primary placenta. This process affected the early development of the embryonic cardiovascular system in mice. Moreover, in vitro experiments showed that PIBF1 promotes the development of cardiovascular characteristics in heart organoids. Our findings show how SynTs organize the barrier and imply their possible roles in supporting embryogenesis, including cardiovascular development. SynT-derived factors and SynT within the placenta may play critical roles in ensuring proper organogenesis of other organs in the embryo.


Asunto(s)
Sistema Cardiovascular , Placenta , Proteínas Gestacionales , Animales , Femenino , Humanos , Ratones , Embarazo , Diferenciación Celular , Desarrollo Embrionario , Placenta/metabolismo , Placentación/fisiología , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Factores Supresores Inmunológicos/metabolismo , Trofoblastos/metabolismo , Sistema Cardiovascular/embriología
15.
Gastroenterology ; 142(7): 1526-35.e6, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22387392

RESUMEN

BACKGROUND & AIMS: Patients with autoimmune hepatitis (AIH) have reduced numbers and function of CD4+CD25(high)FOXP3+ T regulatory cells (Tregs). Tregs can be generated from CD25⁻ (ngTreg) cells, which suppress the immune response less efficiently than Tregs. We investigated whether their differentiation into T-helper (Th)17 cells, an effector subset that has the same CD4+ progenitors as Tregs, accounts for the reduced suppressive functions of ngTregs. We investigated whether blocking interleukin (IL)-17 increased the immunosuppressive activity of Tregs. METHODS: ngTregs were generated from 36 patients with AIH and 23 healthy subjects (controls). During Treg differentiation, expression of IL-17 was inhibited by physical removal of IL-17-secreting cells, exposure to recombinant transforming growth factor ß or neutralizing antibodies against IL-6 and IL-1ß (to promote differentiation of ngTregs vs Th17 cells), small inhibitory RNAs specific for the Th17 transcription factor RORC, or a combination of all these approaches. RESULTS: ngTregs from patients with AIH contained greater proportions of IL-17+ and RORC+ cells than Tregs from controls. All approaches to inhibit IL-17 increased expression of FOXP3 by ngTregs and their suppressive functions. Inhibition of IL-17 led to development of ngTregs that were phenotypically stable and did not acquire proinflammatory properties after exposure to IL-6 and IL-1ß. CONCLUSIONS: Blocking Th17 allows ngTregs to differentiate into functionally stable immune inhibitory cells; this approach might be developed for therapy of patients with AIH.


Asunto(s)
Diferenciación Celular , Hepatitis Autoinmune/inmunología , Interleucina-17/metabolismo , Subunidad alfa del Receptor de Interleucina-2/análisis , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Anticuerpos Neutralizantes/farmacología , Diferenciación Celular/inmunología , Proliferación Celular , Niño , Femenino , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Hepatitis Autoinmune/metabolismo , Humanos , Inmunofenotipificación , Interleucina-17/antagonistas & inhibidores , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Masculino , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Factores Supresores Inmunológicos/metabolismo , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Factor de Crecimiento Transformador beta1/farmacología
16.
Surg Today ; 43(1): 48-54, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23001546

RESUMEN

BACKGROUND AND PURPOSE: Ghrelin, a stomach-derived hormone, stimulates growth hormone secretion and appetite, and inhibits excessive inflammatory response. Plasma ghrelin might affect the inflammatory response to stressful surgical interventions. The aim of this study was to investigate the relationship between serial changes in plasma ghrelin concentrations and the postoperative clinical course after esophagectomy. METHODS: The prospective cohort study subjects were 20 patients with esophageal cancer, who underwent esophagectomy with gastric tube reconstruction. Blood samples were taken six times perioperatively during the course of esophagectomy. RESULTS: The plasma ghrelin level decreased to 33 % (range 15-90 %) on postoperative day (POD) 1, relative to the preoperative level, then recovered to about 50 % by POD 3-10. The duration of systemic inflammatory response syndrome (SIRS) was significantly longer in patients with a marked ghrelin reduction to <33 % on POD 1, than in those with less marked reduction of ≥ 33 % (6.1 ± 1.3 vs. 2.1 ± 0.6 days, P = 0.019). On POD 1, the only inflammatory marker that correlated with the duration of SIRS was the % ghrelin, whereas C-reactive protein, leukocyte count, and IL-6 did not. CONCLUSION: An early postoperative drop in plasma ghrelin correlated with prolonged SIRS after esophagectomy. Thus, the supplementation of low plasma ghrelin may help minimize excess inflammatory response in these patients.


Asunto(s)
Carcinoma de Células Escamosas/cirugía , Neoplasias Esofágicas/cirugía , Esofagectomía , Ghrelina/sangre , Ghrelina/fisiología , Complicaciones Posoperatorias/diagnóstico , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Periodo Posoperatorio , Estudios Prospectivos , Factores Supresores Inmunológicos/metabolismo , Factores de Tiempo , Adulto Joven
17.
Clin Oral Investig ; 17(5): 1365-73, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22893035

RESUMEN

OBJECTIVES: We sought to investigate the expression of cells with immunosuppressive/protumorigenic phenotypes in oral lichen planus (OLP), such as M2-tumor-associated macrophages (TAM2), myeloid-derived suppressive cells (MDSCs), and regulatory T cells (Tregs) in association with clinical parameters. MATERIALS AND METHODS: Cases of hyperkeratotic (HK)-OLP (n = 23) and erosive (E)-OLP (n = 26) were immunohistochemically stained to determine the percentages of CD163-TAM2, CD80-MDSCs, and FOXP3-Tregs of proinflammatory CD121a-Th17, CD4 and CD8 lymphocytes, and of cells positive for nuclear factor kappa B (NF-κB) and transforming growth factor beta. Clinical parameters included symptoms, treatment approach, treatment response, and others. RESULTS: The inflammatory infiltrate in HK-OLP and E-OLP contained immunosuppressive cells; however, their pattern of expression was compatible with a proinflammatory response [membranous CD163-TAM2 staining (not extracellular), CD80+ lymphocytes (not macrophages), and a few Tregs]. The presence of CD4+, CD8+, and CD121a+ T lymphocytes was extensive. TAM2 were more frequent in E-OLP than in HK-OLP (P = 0.017). A higher frequency of CD80+ lymphocytes was associated with partial to no response to treatment (P = 0.028). Nuclear expression of NF-κB in the inflammatory cells was absent. CONCLUSIONS: The pattern of expression of the immunosuppressive cells, together with numerous CD4+, CD8+, and Th17-CD121a+ lymphocytes, suggest an extensive proinflammatory response rather than an immunosuppressive/protumorigenic response. CLINICAL RELEVANCE: The frequency of selective types of inflammatory cells calls for individual profile analyses of inflammatory infiltrates and individually adjusted treatment.


Asunto(s)
Tolerancia Inmunológica , Inflamación/inmunología , Liquen Plano Oral/inmunología , Liquen Plano Oral/patología , Adulto , Anciano , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Antígeno B7-1/inmunología , Femenino , Humanos , Macrófagos , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Fenotipo , Receptores de Superficie Celular/inmunología , Factores Supresores Inmunológicos/metabolismo , Linfocitos T Citotóxicos , Linfocitos T Colaboradores-Inductores , Linfocitos T Reguladores , Células Th17 , Factor de Crecimiento Transformador beta/inmunología
18.
Yi Chuan ; 35(12): 1377-83, 2013 Dec.
Artículo en Zh | MEDLINE | ID: mdl-24645347

RESUMEN

MNSFbeta (Monoclonal nonspecific suppressor factor beta) is a natural immunosuppressive factor which has been reported to be involved in various biological processes, such as immune responses, cell division, stress response, cell apoptosis, and nuclear transport. However, study on porcine MNSFbeta has been rarely reported. In this study, the full-length sequence of porcine MNSFbeta (GenBank accession number: KF77642500) was predicted in silicon and its cDNA sequence was obtained through RT-PCR from porcine spleen. The nucleic acid and protein sequences were analyzed. Then, the gene was subcloned into pEGFP-C1 to construct a recombinant plasmid pEGFP-MNSFbeta which was transfected into swine umbilical vein endothelial cells (SUVECs) using Lipofectamine 2000. The expression of GFP was detected by fluorescence microscopy, Western blot, and laser confocal fluorescence microscopy. The spatial expression patterns of porcine MNSFbeta were detected by real-time qPCR. Results showed that the full length of porcine MNSFbeta was 402 bp encoding 133 amino acids with only one exon. Bioinformatics analysis showed that porcine MNSFbeta protein was a stable protein consisting of a ubiquitin-like domain fused to the ribosomal protein S30 with no signal peptide. The analyses of homology and phylogenetic tree of porcine MNSFbeta and its homologs in other 18 species showed that the identities of MNSFbeta protein sequence were higher than 91% among different species and the evolutionary distance was less than 0.05. It indicates that MNSFbeta is highly conserved in the process of evolution. Fluorescence signal showed that the fusion protein GFP-MNSFbeta was successfully expressed in SUVECs which was then confirmed by Western blot. Laser confocal fluorescence microscopy showed that MNSFbeta was expressed in both nucleus and cytoplasm. Analysis of spatial expression patterns showed that procine MNSFbeta was widely expressed in immune tissues, but not in lung, suggesting that MNSFbeta may play an important role in immune response.


Asunto(s)
Factores Supresores Inmunológicos/metabolismo , Animales , Western Blotting , Biología Computacional , Masculino , Estructura Secundaria de Proteína , Factores Supresores Inmunológicos/química , Factores Supresores Inmunológicos/genética , Porcinos
19.
Am J Reprod Immunol ; 90(2): e13745, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37491933

RESUMEN

PROBLEM: Recurrent pregnancy loss (RPL) is the spontaneous loss of two or more consecutive pregnancies prior to 20 weeks of gestation, occurring in 1% of the reproductive-age population. It is a major cause of infertility in India with a staggering 7.46% prevalence rate. METHOD OF STUDY: Blood and product of conception (POCs) from RPL cases (n = 65) were enrolled for this study, along with cases of medically terminated pregnancy (MTP, n = 80) and term delivery cases (n = 90) as control. ELISA for progesterone and progesterone induced blocking factor (PIBF) levels was carried out, followed by mRNA expression analysis of progesterone receptor isoform B (PR-B) and its downstream immunomodulatory effectors, namely, PIBF, IL-10 and IL-12. Screening of PROGINS haplotype of PR gene and PIBF polymorphism were also conducted to correlate with their respective gene expression profiles. RESULTS: Serum progesterone level was found to be comparable in the RPL and MTP cases. Although the mRNA expression of PR-B was found to be downregulated in the RPL cases, no significant PROGINS haplotype was observed. Presence of a single nucleotide polymorphism (SNP) in the PIBF gene (rs1372000) was more in healthy controls compared to RPL cases. Serum PIBF levels were found to be lower in the RPL cases with a resultant increase in IL-12 and a decrease in IL-10 mRNA expression in these cases. CONCLUSIONS: This study indicates that progesterone, acting through PIBF, modulates the immunological state of pregnancy to be Th1-biased in RPL, indicative of a pro-inflammatory, labour-like state not desired for a healthy pregnancy.


Asunto(s)
Aborto Habitual , Progesterona , Embarazo , Femenino , Humanos , Progesterona/farmacología , Citocinas , Interleucina-10/genética , Aborto Habitual/genética , Interleucina-12 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores Supresores Inmunológicos/genética , Factores Supresores Inmunológicos/metabolismo
20.
Mol Cell Biochem ; 364(1-2): 39-43, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22273981

RESUMEN

Post-translational modification by monoclonal nonspecific suppressor factor ß (MNSFß) has been involved in the regulation of a variety of cellular processes. Previous studies have demonstrated that MNSFß covalently binds to the intracellular pro-apoptotic protein Bcl-G and regulates TLR-4-mediated signal transduction. Recently, we found that MNSFß also covalently conjugates to endophilin II, a member of the endophilin A family, and inhibits the signal pathway upstream of IKK activation, but not downstream of TLR-2 signaling. In this study, we further examined the mechanism of action of MNSFß in TLR-2-mediated signal transduction in macrophage-like cell line Raw264.7 cells. Although MNSFß siRNA enhanced Pam(3)CDK(4) (TLR-2-specific ligand)-stimulated TNFα production, Bcl-G siRNA did not affect. MNSFß cDNA inhibited the Pam(3)CDK(4)-stimulated TNFα production. High-molecular weight (130 kDa) MNSFß-adduct was induced in Pam(3)CDK(4)-stimulated Raw264.7 cells. This MNSFß-adduct was not induced by LPS, indicative of the specificity of TLR-2-mediated signal transduction. Similar observations were seen in BALB/c peritoneal macrophages. Interestingly, 40-kDa MNSFß-adduct was tyrosine phosphorylated by Pam(3)CDK(4) stimulation. Collectively, novel MNSFß-adducts may regulate TLR-2 signaling pathway in macrophages.


Asunto(s)
Regulación de la Expresión Génica/genética , Factores Supresores Inmunológicos/metabolismo , Receptor Toll-Like 2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño , Transducción de Señal/genética , Factores Supresores Inmunológicos/genética , Receptor Toll-Like 2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA