Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Neuroinflammation ; 21(1): 79, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38549144

RESUMEN

Stimulation of the inflammatory reflex (IR) is a promising strategy for treating systemic inflammatory disorders. Recent studies suggest oral sodium bicarbonate (NaHCO3) as a potential activator of the IR, offering a safe and cost-effective treatment approach. However, the mechanisms underlying NaHCO3-induced anti-inflammatory effects remain unclear. We investigated whether oral NaHCO3's immunomodulatory effects are mediated by the splenic nerve. Female rats received NaHCO3 or water (H2O) for four days, and splenic immune markers were assessed using flow cytometry. NaHCO3 led to a significant increase (p < 0.05, and/or partial eta squared > 0.06) in anti-inflammatory markers, including CD11bc + CD206 + (M2-like) macrophages, CD3 + CD4 + FoxP3 + cells (Tregs), and Tregs/M1-like ratio. Conversely, proinflammatory markers, such as CD11bc + CD38 + TNFα + (M1-like) macrophages, M1-like/M2-like ratio, and SSChigh/SSClow ratio of FSChighCD11bc + cells, decreased in the spleen following NaHCO3 administration. These effects were abolished in spleen-denervated rats, suggesting the necessity of the splenic nerve in mediating NaHCO3-induced immunomodulation. Artificial neural networks accurately classified NaHCO3 and H2O treatment in sham rats but failed in spleen-denervated rats, highlighting the splenic nerve's critical role. Additionally, spleen denervation independently influenced Tregs, M2-like macrophages, Tregs/M1-like ratio, and CD11bc + CD38 + cells, indicating distinct effects from both surgery and treatment. Principal component analysis (PCA) further supported the separate effects. Our findings suggest that the splenic nerve transmits oral NaHCO3-induced immunomodulatory changes to the spleen, emphasizing NaHCO3's potential as an IR activator with therapeutic implications for a wide spectrum of systemic inflammatory conditions.


Asunto(s)
Bazo , Nervio Vago , Ratas , Femenino , Animales , Antiinflamatorios/farmacología , Inmunomodulación , Macrófagos
2.
Pharmacol Res ; 187: 106525, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36441036

RESUMEN

Stimulation of the inflammatory reflex (IR) is a promising strategy to treat systemic inflammatory disorders. However, this strategy is hindered by the cost and side effects of traditional IR activators. Recently, oral intake of sodium bicarbonate (NaHCO3) has been suggested to activate the IR, providing a safe and inexpensive alternative. Critically, the mechanisms whereby NaHCO3 might achieve this effect and more broadly the pathways underlying the IR remain poorly understood. Here, we argue that the recognition of NaHCO3 as a potential IR activator presents exciting clinical and research opportunities. To aid this quest, we provide an integrative review of our current knowledge of the neural and cellular pathways mediating the IR and discuss the status of physiological models of IR activation. From this vantage point, we derive testable hypotheses on potential mechanisms whereby NaHCO3 might stimulate the IR and compare NaHCO3 with classic IR activators. Elucidation of these mechanisms will help determine the therapeutic value of NaHCO3 as an IR activator and provide new insights into the IR circuitry.


Asunto(s)
Reflejo , Reflejo/fisiología
3.
Biomedicines ; 12(6)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38927570

RESUMEN

Protein kinase C delta (PKCδ) has emerged as a key protective molecule against systemic lupus erythematosus (SLE or lupus), an autoimmune disease characterized by anti-double stranded (ds) DNA IgGs. Although PKCδ-deficient mice and lupus patients with mutated PRKCD genes clearly demonstrate the requirement for PKCδ in preventing lupus autoimmunity, this critical tolerance mechanism remains poorly understood. We recently reported that PKCδ acts as a key regulator of B cell tolerance by selectively deleting anti-dsDNA B cells in the germinal center (GC). PKCδ's tolerance function is activated by sphingomyelin synthase 2 (SMS2), a lipid enzyme whose expression is generally reduced in B cells from lupus patients. Moreover, pharmacologic strengthening of the SMS2/PKCδ tolerance pathway alleviated lupus pathogenesis in mice. Here, we review relevant publications in order to provide mechanistic insights into PKCδ's tolerance activity and discuss the potential significance of therapeutically targeting PKCδ's tolerance activity in the GC for selectively inhibiting lupus autoimmunity.

4.
Front Immunol ; 15: 1339325, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38444862

RESUMEN

Introduction: The microphthalmia transcription factor Mitf has been shown to regulate B cell activation and tolerance. However, the underlying B cell-specific mechanisms responsible, and those that distinguish Mitf from closely related Mitf/TFE (MiT) transcription factors Tfe3, Tfeb, and Tfec, remain obscure. Methods: Two complementary mouse models of Mitf and MiT deficiency were used: the Mitfmi-vga9/mi-vga9 systemic loss-of-function mutation, and B-cell specific MiT family inactivation via transgenic expression of a trans-dominant negative (TDN) protein (TDN-B). These models were employed to identify MiT family candidate target genes and pathways. Results: Both models displayed spontaneous splenomegaly coincident with elevated plasma cell numbers, autoantibody titers, and proteinuria. These abnormalities appeared dependent on T helper cells, but independent of other non-B cell intrinsic effects of systemic Mitf inactivation. MiT inactivation in B cells augmented aspects of lupus-like autoimmune disease on the C57BL/6-Faslpr/lpr background. In both models, RNAseq of ex vivo resting B cells showed transcriptional upregulation of genes that control cell cycle, germinal center responses, and plasma cell differentiation. Among the genes strongly upregulated in both models were Socs6, Isp53 (Baiap1), S1pR2, and IgG2b/c. Mitf null B cells, but not TDN-B cells, showed evidence of type I interferon dysregulation. Discussion: These studies clarify Mitf's role as 1) a key regulator of a B cell intrinsic germinal center program that influences self-tolerance through novel target genes, and 2) a regulator of systemic inflammatory processes that can impact the B cell microenvironment. This distinction of Mitf's function from that of related MiT transcription factors advances our understanding of B cell regulation and autoimmunity.


Asunto(s)
Linfocitos B , Centro Germinal , Animales , Ratones , Expresión Génica , Homeostasis , Ratones Endogámicos C57BL
5.
STAR Protoc ; 3(2): 101388, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35600926

RESUMEN

Highly enriched germinal center (GC) B cell populations are essential for studying humoral immunity. Current MACS protocols that isolate untouched GC B cells require GC induction and typically require further FACS purification with direct antibody labeling to achieve sufficiently high purities. We present a MACS protocol with progressive and repeated negative selections that yields highly purified untouched GC B cells from both unimmunized and GC-induced mice and allows further FACS isolation of unlabeled GC B cells from remaining debris by scatter.


Asunto(s)
Linfocitos B , Centro Germinal , Animales , Anticuerpos , Inmunidad Humoral , Recuento de Linfocitos , Ratones
6.
Cell Rep ; 36(9): 109624, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469734

RESUMEN

B cell tolerance prevents autoimmunity by deleting or deactivating autoreactive B cells that otherwise may cause autoantibody-driven disorders, including systemic lupus erythematosus (lupus). Lupus is characterized by immunoglobulin Gs carrying a double-stranded (ds)-DNA autospecificity derived mainly from somatic hypermutation in the germinal center (GC), pointing to a checkpoint breach of GC B cell tolerance that leads to lupus. However, tolerance mechanisms in the GC remain poorly understood. Here, we show that upregulated sphingomyelin synthase 2 (SMS2) in anti-dsDNA GC B cells induces apoptosis by directly activating protein kinase C δ (PKCδ)'s pro-apoptotic activity. This tolerance mechanism prevents lupus autoimmunity in C57/BL6 mice and can be stimulated pharmacologically to inhibit lupus pathogenesis in lupus-prone NZBWF1 mice. Patients with lupus consistently have substantially reduced SMS2 expression in B cells and to an even greater extent in autoimmune-prone, age-associated B cells, suggesting that patients with lupus have insufficient SMS2-regulated B cell tolerance.


Asunto(s)
Autoinmunidad , Linfocitos B/enzimología , Centro Germinal/enzimología , Tolerancia Inmunológica , Lupus Eritematoso Sistémico/enzimología , Proteína Quinasa C-delta/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/deficiencia , Animales , Apoptosis , Autoinmunidad/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Células Cultivadas , Modelos Animales de Enfermedad , Activación Enzimática , Activadores de Enzimas/farmacología , Femenino , Predisposición Genética a la Enfermedad , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Centro Germinal/patología , Tolerancia Inmunológica/efectos de los fármacos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/prevención & control , Ratones Endogámicos C57BL , Ratones Endogámicos NZB , Ratones Noqueados , Proteína Quinasa C-delta/genética , Transducción de Señal , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo
7.
J Immunol ; 181(6): 4098-106, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18768866

RESUMEN

The truncated/V(H)-less mouse H chain Dmu forms precursor B cell receptors with the surrogate L chain complex that promotes allelic exclusion but not other aspects of pre-B cell development, causing most progenitor B cells expressing this H chain to be eliminated at the pre-B cell checkpoint. However, there is evidence that Dmu-lambda1 complexes can be made and are positively selected during fetal life but cannot sustain adult B lymphopoiesis. How surrogate and conventional L chains interpret Dmu's unusual structure and how that affects signaling outcome are unclear. Using nonlymphoid and primary mouse B cells, we show that secretion-competent lambda1 L chains could associate with both full-length H chains and Dmu, whereas secretion-incompetent lambda1 L chains could only do so with full-length H chains. In contrast, Dmu could not form receptors with a panel of kappa L chains irrespective of their secretion properties. This was due to an incompatibility of Dmu with the kappa-joining and constant regions. Finally, the Dmu-lambda1 receptor was less active than the full-length mouse mu-lambda1 receptor in promoting growth under conditions of limiting IL-7. Thus, multiple receptor-dependent mechanisms operating at all stages of B cell development limit the contribution of B cells with Dmu H chain alleles to the repertoire.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Diferenciación Celular/inmunología , Reordenamiento Génico de Cadena Ligera de Linfocito B , Cadenas Pesadas de Inmunoglobulina/fisiología , Inmunoglobulina de Cadenas Ligeras Subrogadas/fisiología , Cadenas kappa de Inmunoglobulina/fisiología , Cadenas lambda de Inmunoglobulina/fisiología , Células Madre/inmunología , Alelos , Animales , Subgrupos de Linfocitos B/metabolismo , Diferenciación Celular/genética , Línea Celular , Línea Celular Transformada , Células Cultivadas , Humanos , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/genética , Inmunoglobulina de Cadenas Ligeras Subrogadas/biosíntesis , Inmunoglobulina de Cadenas Ligeras Subrogadas/genética , Cadenas kappa de Inmunoglobulina/biosíntesis , Cadenas kappa de Inmunoglobulina/genética , Cadenas lambda de Inmunoglobulina/biosíntesis , Cadenas lambda de Inmunoglobulina/genética , Ratones , Ratones Noqueados , Mutagénesis , Receptores de Antígenos de Linfocitos B/biosíntesis , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/fisiología , Células Madre/metabolismo
8.
J Investig Med ; 65(4): 754-758, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28104820

RESUMEN

The use of immune checkpoint inhibitors to treat malignant tumors with microsatellite instability is an emerging new modality. This is based on the observations that these tumors may have a high mutation rate-thus a potential source of tumor-specific neoantigens-and harbor infiltrating cytotoxic T cells in response, suggesting that they may be particularly susceptible to immune checkpoint therapy. PUBMED and ASCO library were systematically reviewed to identify all relevant data that involved the use of immune checkpoint inhibitors in the treatment of cancers with microsatellite instability. The manual search retrieved a total of 3 relevant articles and 1 abstract published between 2015 and 2016. A total of 61 patients with colorectal, 3 with ampullary/cholangiocarcinoma, 2 with endometrial carcinomas, 3 with small bowel cancers, 2 with glioblastoma multiforme, and 1 with bladder cancer with reported efficacy results were reviewed. All the patients had stage IV cancer and were treated with immune checkpoint inhibitors until progression of disease or intolerable side effects emerged. The range of objective response rate was 25-71%. Responses were also durable with progression-free survival at 20 weeks of around 67-78% and to 46% at 1 year. The use of immune checkpoint inhibitors is effective in cancers that express microsatellite instability.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Sistema Inmunológico/patología , Neoplasias/inmunología , Neoplasias/patología , Humanos , Inestabilidad de Microsatélites , Neoplasias/genética
9.
Cell Rep ; 16(10): 2630-2640, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27568564

RESUMEN

Endoplasmic reticulum (ER)-associated degradation (ERAD) is a principal mechanism that targets ER-associated proteins for cytosolic proteasomal degradation. Here, our data demonstrate a critical role for the Sel1L-Hrd1 complex, the most conserved branch of ERAD, in early B cell development. Loss of Sel1L-Hrd1 ERAD in B cell precursors leads to a severe developmental block at the transition from large to small pre-B cells. Mechanistically, we show that Sel1L-Hrd1 ERAD selectively recognizes and targets the pre-B cell receptor (pre-BCR) for proteasomal degradation in a BiP-dependent manner. The pre-BCR complex accumulates both intracellularly and at the cell surface in Sel1L-deficient pre-B cells, leading to persistent pre-BCR signaling and pre-B cell proliferation. This study thus implicates ERAD mediated by Sel1L-Hrd1 as a key regulator of B cell development and reveals the molecular mechanism underpinning the transient nature of pre-BCR signaling.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Puntos de Control del Ciclo Celular , Degradación Asociada con el Retículo Endoplásmico , Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Antígenos CD19/metabolismo , Ciclo Celular , Tamaño de la Célula , Péptidos y Proteínas de Señalización Intracelular , Ratones Endogámicos C57BL , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/patología , Receptores de Antígenos de Linfocitos B , Especificidad por Sustrato , Factor de Transcripción CHOP/metabolismo
10.
Mol Immunol ; 47(6): 1195-206, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20116104

RESUMEN

PreBCR signaling is critical for B cell development and normally depends on the association of a nascent, component Ig H chain with the surrogate L chain (SLC), which helps ensure that only B cells that synthesize structurally sound antibody can develop. How the invariant and lambda-like SLC vets billions of unique V(H) domains for compatibility with polymorphic kappa and lambda L chains is unclear, because the SLC is composed of not only the Ig domains of VpreB and lambda5, but also the unique regions (URs) that reside at what would be the L chain CDR3. We evaluated the contribution of the Ig and UR domains of lambda5 to H chain screening by evaluating the preBCR-forming capability of lambda5 mutants with a diverse panel of H chains. Using transformed mouse B cells, we demonstrate that the Ig domain of lambda5 was sufficient and its UR dispensable for the rejection of V(H)Q52 and V(H)10 SLC-incompatible H chains. In contrast, the lambda5 UR was necessary to discriminate between SLC-incompatible and -compatible V(H)81X H chains. Substituting the Ig domains of lambda5 with equivalent kappa sequences impaired the SLC's ability to escort all H chains to the surface. Two SLC-incompatible H chains were able to form surface BCRs with two kappa L chains, indicating that the SLC's ability to predict the L chain compatibility of a H chain is not absolute. In sum, lambda5 differentially relies on the lambda-like Ig and UR to probe H chain structure to best accommodate diversity among H chains.


Asunto(s)
Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/inmunología , Inmunoglobulina de Cadenas Ligeras Subrogadas/química , Inmunoglobulina de Cadenas Ligeras Subrogadas/inmunología , Alelos , Secuencia de Aminoácidos , Animales , Línea Celular , Humanos , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/inmunología , Cadenas kappa de Inmunoglobulina/inmunología , Ratones , Datos de Secuencia Molecular , Receptores de Células Precursoras de Linfocitos B/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido
11.
Mol Endocrinol ; 24(8): 1512-28, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20573688

RESUMEN

Cervical ripening during pregnancy is a profound change in cervix structure and function characterized by increases in the proinflammatory cytokine IL-8 and dissolution of the cervical extracellular matrix. Relatively little is known about the molecular mechanisms that underlie these events. Here, we report identification of a novel isoform of micropthalmia-associated transcription factor in human cervical stromal cells (MiTF-CX) that is down-regulated 12-fold during cervical ripening and that represses expression of IL-8. Ectopic expression of MiTF-CX in human cervical stromal cells resulted in substantial suppression of endogenous IL-8 mRNA and protein expression, whereas expression of dominant negative MiTF-CX mutants with impaired DNA binding resulted in dramatic increases in IL-8 production. Gel shift, reporter gene, and chromatin immunoprecipitation assays revealed one strong binding site (E-box (-397) CACATG(-391)) in the human IL-8 promoter that was crucial for mediating transcriptional repression by MiTF-CX. Moreover, we show that MiTF-CX expression in the cervix was itself positively autoregulated via two E-box motifs within a 2.1-kb promoter fragment. We therefore propose that maintenance of cervical competency during pregnancy is an active process maintained through suppression of IL-8 by the transcription factor MiTF-CX. During cervical ripening, loss of MiTF-CX would result in significant up-regulation of IL-8 mRNA and protein synthesis, thereby leading to recruitment and activation of leukocytes within the cervix and dissolution of the extracellular matrix.


Asunto(s)
Cuello del Útero/citología , Interleucina-8/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Isoformas de Proteínas/metabolismo , Células del Estroma/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ratones , Factor de Transcripción Asociado a Microftalmía/genética , Parto/genética , Reacción en Cadena de la Polimerasa , Embarazo , Isoformas de Proteínas/genética
12.
J Immunol ; 176(11): 6862-72, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16709846

RESUMEN

Signals from the precursor BCR (preBCR) cause proliferation and differentiation of progenitor (pro-) B cells into pre-B cells. Given the very low amounts of surface preBCRs and the demonstrated cell autonomy of preBCR signaling, we examined the possible occurrence of preBCR signal propagation from intracellular membranes such as the endoplasmic reticulum (ER) and the trans-Golgi network (TGN) in transformed and primary pro-B cells. PreBCRs composed of normal Ig mu or truncated Dmu heavy chains (HCs) were redirected to intracellular sites via localization sequences appended to the HC cytoplasmic tail. PreBCR complexes retained in the TGN or shunted from the TGN to lysosomes were as or 50% as active as the corresponding wild-type preBCRs in directing preBCR-dependent events, including CD2 and CD22 expression and proliferation in primary pro-B cells. This occurred despite their low to undetectable surface expression in transformed cells, which otherwise allowed significant surface accumulation of wild-type preBCRs. In contrast, ER-retained preBCRs were inactive. These results suggest that preBCR signaling is remarkably tolerant of dramatic changes in its subcellular distribution within post-ER compartments and support the possibility that the preBCR can activate signaling pathways in the TGN as well as the plasma membrane.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Membrana Celular/inmunología , Membranas Intracelulares/inmunología , Glicoproteínas de Membrana/fisiología , Transducción de Señal/inmunología , Células Madre/inmunología , Secuencia de Aminoácidos , Animales , Subgrupos de Linfocitos B/metabolismo , Línea Celular , Línea Celular Transformada , Membrana Celular/metabolismo , Células Cultivadas , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Receptores de Células Precursoras de Linfocitos B , Receptores de Antígenos de Linfocitos B , Células Madre/metabolismo
13.
Nat Immunol ; 7(10): 1082-91, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16936731

RESUMEN

TFE3 and TFEB are broadly expressed transcription factors related to the transcription factor Mitf. Although they have been linked to cytokine signaling pathways in nonlymphoid cells, their function in T cells is unknown. TFE3-deficient mice are phenotypically normal, whereas TFEB deficiency causes early embryonic death. We now show that combined inactivation of TFE3 and TFEB in T cells resulted in a hyper-immunoglobulin M syndrome due to impaired expression of CD40 ligand by CD4(+) T cells. Native TFE3 and TFEB bound to multiple cognate sites in the promoter of the gene encoding CD40 ligand (Cd40lg), and maximum Cd40lg promoter activity and gene expression required TFE3 or TFEB. Thus, TFE3 and TFEB are direct, physiological and mutually redundant activators of Cd40lg expression in activated CD4(+) T cells critical for T cell-dependent antibody responses.


Asunto(s)
Formación de Anticuerpos/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Ligando de CD40/genética , Regulación de la Expresión Génica , Síndrome de Inmunodeficiencia con Hiper-IgM/genética , Timo/inmunología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/agonistas , Ligando de CD40/análisis , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas/genética
14.
J Immunol ; 175(1): 358-66, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15972669

RESUMEN

Signals transduced by precursor-BCRs (pre-BCRs) composed of Ig mu heavy chains (HCs) and the surrogate L chain components lambda5 and VpreB are critical for B cell development. A conserved unique region (UR) of lambda5 was shown to activate pre-BCR complexes in transformed cells and to engage putative ligands, but its contribution to pre-B cell development is not known. It is also not clear why the lambda-like sequences in lambda5 are used to select HCs that will associate mainly with kappa L chains. In this study, we show that, in transformed and primary mouse B cell progenitors, receptors containing full-length HCs and lacking the lambda5UR were expressed at higher surface levels, but exhibited reduced activity compared with normal pre-BCRs in supporting developmental changes that accompany the progenitor to pre-B cell transition in primary cell culture systems and in the bone marrow in vivo. In contrast, deletion of the lambda5UR did not change net signaling output by the Dmu-pre-BCR, a developmentally defective receptor that exhibited impaired activity in the primary cell culture system. Moreover, the lambda-like sequences in lambda5 were more accommodating than kappa in supporting surface expression and signaling by the different HCs. These results show that the lambda5UR is important, although not essential, for surrogate L chain-dependent receptor signaling in primary cells, and furthermore may help allow discrimination of signaling competency between normal and Dmu-pre-BCRs. That the lambda-like portion of lambda5 in the absence of the UR was nondiscriminatory suggests that the lambda5UR focuses pre-BCR-dependent selection on the HC V region.


Asunto(s)
Linfocitos B/inmunología , Cadenas Ligeras de Inmunoglobulina/química , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Linfocitos B/citología , Transformación Celular Viral , Células Cultivadas , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis , Humanos , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/metabolismo , Inmunoglobulina de Cadenas Ligeras Subrogadas , Técnicas In Vitro , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Estructura Terciaria de Proteína , Transducción de Señal
15.
J Biol Chem ; 280(34): 30225-35, 2005 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-15994295

RESUMEN

Translocations of the genes encoding the related transcription factors TFE3 and TFEB are almost exclusively associated with a rare juvenile subset of renal cell carcinoma and lead to overexpression of TFE3 or TFEB protein sequences. A better understanding of how deregulated TFE3 and TFEB contribute to the transformation process requires elucidating more of the normal cellular processes in which they participate. Here we identify TFE3 and TFEB as cell type-specific leukemia inhibitory factor-responsive activators of E-cadherin. Overexpression of TFE3 or TFEB in 3T3 cells activated endogenous and reporter E-cadherin expression. Conversely, endogenous TFE3 and/or TFEB was required for endogenous E-cadherin expression in primary mouse embryonic fibroblasts and human embryonic kidney cells. Chromatin precipitation analyses and E-cadherin promoter reporter gene assays revealed that E-cadherin induction by TFE3 or TFEB was primarily or exclusively direct and mitogen-activated protein kinase-dependent in those cell types. In mouse embryonic fibroblasts, TFE3 and TFEB activation of E-cadherin was responsive to leukemia inhibitory factor. In 3T3 cells, TFE3 and TFEB expression also induced expression of Wilms' tumor-1, another E-cadherin activator. In contrast, E-cadherin expression in model mouse and canine renal epithelial cell lines was indifferent to inhibition of endogenous TFE3 and/or TFEB and was reduced by TFE3 or TFEB overexpression. These results reveal new cell type-specific activities of TFE3 and TFEB which may be affected by their mutation.


Asunto(s)
Cadherinas/metabolismo , Carcinoma/metabolismo , Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica , Interleucina-6/metabolismo , Neoplasias Renales/metabolismo , Proteínas de Neoplasias/fisiología , Factores de Transcripción/fisiología , Proteínas WT1/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Western Blotting , Línea Celular , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , ADN Complementario/metabolismo , Perros , Activación Enzimática , Células Epiteliales , Fibroblastos/metabolismo , Genes Reporteros , Vectores Genéticos , Humanos , Inmunoprecipitación , Riñón/metabolismo , Factor Inhibidor de Leucemia , Luciferasas/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Microscopía Confocal , Mutación , Células 3T3 NIH , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , Retroviridae/genética , Retroviridae/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Transfección
16.
J Immunol ; 169(12): 6900-9, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12471123

RESUMEN

Adapter molecules that promote protein-protein interactions play a central role in T lymphocyte differentiation and activation. In this study, we examined the role of the T lymphocyte-expressed adapter protein and Src kinase substrate, Sin, on thymocyte function using transgenic mice expressing an activated, truncated allele of Sin (SinDeltaC). We found that SinDeltaC expression led to reduced numbers of CD4(+) and CD8(+) single-positive cells and reduced thymic cellularity due to increased thymocyte apoptosis. Because the adapter properties of Sin are mediated by tyrosine-based motifs and given that Sin is a substrate for Src tyrosine kinases, we examined the involvement of these kinases in the inhibitory effects of SinDeltaC. We found that in transgenic thymocytes, SinDeltaC was constitutively phosphorylated by the Src kinase Fyn, but not by the related kinase Lck. Using SinDeltaC and fyn(-/-) animals, we also found that the expression of Fyn was required for the inhibitory effect of SinDeltaC on thymocyte apoptosis but not for SinDeltaC-mediated inhibition of T cell maturation. The inhibitory effect of SinDeltaC on thymocyte maturation correlated with defective activation of the mitogen-activated protein kinase extracellular signal-regulated kinase. Our results suggest that the Sin mutant inhibits thymocyte differentiation through Fyn-dependent and -independent mechanisms and that endogenous Sin may be an important regulator of thymocyte development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Portadoras/genética , Proteínas de la Membrana , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas/genética , Linfocitos T/metabolismo , Timo/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Animales , Apoptosis/genética , Apoptosis/inmunología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/fisiología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/fisiología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfoproteínas/biosíntesis , Fosforilación , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-fyn , Receptores de Antígenos de Linfocitos T/metabolismo , Eliminación de Secuencia , Especificidad por Sustrato , Linfocitos T/inmunología , Linfocitos T/patología , Timo/inmunología , Timo/patología , Transgenes/inmunología , Familia-src Quinasas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA