Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(33): e2304943120, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37549290

RESUMEN

Conventional dendritic cells (cDCs) are required for peripheral T cell homeostasis in lymphoid organs, but the molecular mechanism underlying this requirement has remained unclear. We here show that T cell-specific CD47-deficient (Cd47 ΔT) mice have a markedly reduced number of T cells in peripheral tissues. Direct interaction of CD47-deficient T cells with cDCs resulted in activation of the latter cells, which in turn induced necroptosis of the former cells. The deficiency and cell death of T cells in Cd47 ΔT mice required expression of its receptor signal regulatory protein α on cDCs. The development of CD4+ T helper cell-dependent contact hypersensitivity and inhibition of tumor growth by cytotoxic CD8+ T cells were both markedly impaired in Cd47 ΔT mice. CD47 on T cells thus likely prevents their necroptotic cell death initiated by cDCs and thereby promotes T cell survival and function.


Asunto(s)
Antígeno CD47 , Linfocitos T CD8-positivos , Animales , Ratones , Antígeno CD47/genética , Antígeno CD47/metabolismo , Linfocitos T CD8-positivos/metabolismo , Supervivencia Celular , Células Dendríticas/metabolismo , Necroptosis , Receptores Inmunológicos/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34949714

RESUMEN

The interaction of signal regulatory protein α (SIRPα) on macrophages with CD47 on cancer cells is thought to prevent antibody (Ab)-dependent cellular phagocytosis (ADCP) of the latter cells by the former. Blockade of the CD47-SIRPα interaction by Abs to CD47 or to SIRPα, in combination with tumor-targeting Abs such as rituximab, thus inhibits tumor formation by promoting macrophage-mediated ADCP of cancer cells. Here we show that monotherapy with a monoclonal Ab (mAb) to SIRPα that also recognizes SIRPß1 inhibited tumor formation by bladder and mammary cancer cells in mice, with this inhibitory effect being largely dependent on macrophages. The mAb to SIRPα promoted polarization of tumor-infiltrating macrophages toward an antitumorigenic phenotype, resulting in the killing and phagocytosis of cancer cells by the macrophages. Ablation of SIRPα in mice did not prevent the inhibitory effect of the anti-SIRPα mAb on tumor formation or its promotion of the cancer cell-killing activity of macrophages, however. Moreover, knockdown of SIRPß1 in macrophages attenuated the stimulatory effect of the anti-SIRPα mAb on the killing of cancer cells, whereas an mAb specific for SIRPß1 mimicked the effect of the anti-SIRPα mAb. Our results thus suggest that monotherapy with Abs to SIRPα/SIRPß1 induces antitumorigenic macrophages and thereby inhibits tumor growth and that SIRPß1 is a potential target for cancer immunotherapy.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Antineoplásicos/farmacología , Inmunoterapia/métodos , Macrófagos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Antígeno CD47/metabolismo , Línea Celular Tumoral , Ratones , Receptores de Superficie Celular/genética , Receptores Inmunológicos/genética , Rituximab , Resultado del Tratamiento , Vejiga Urinaria
3.
Cancer Sci ; 114(5): 1871-1881, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36788737

RESUMEN

Langerhans cell histiocytosis (LCH) is a rare neoplastic disorder characterized by inflammatory lesions arising from the anomalous accumulation of pathogenic CD1a+ CD207+ dendritic cells (DCs). SIRPα is a transmembrane protein highly expressed in myeloid cells such as DCs and macrophages. Here we show that SIRPα is a potential therapeutic target for LCH. We found that SIRPα is expressed in CD1a+ cells of human LCH lesions as well as in CD11c+ DCs in the spleen, liver, and lung of a mouse model of LCH (BRAFV600ECD11c mouse), in which an LCH-associated active form of human BRAF is expressed in a manner dependent on the mouse Cd11c promoter. BRAFV600ECD11c mice manifested markedly increased numbers of CD4+ T cells, regulatory T cells, and macrophages as well as of CD11c+ MHCII+ DCs in the spleen. Monotherapy with a mAb to SIRPα greatly reduced the percentage of CD11c+ MHCII+ DCs in peripheral blood, LCH-like lesion size in the liver, and the number of CD11c+ MHCII+ DCs in the spleen of the mutant mice. Moreover, this mAb promoted macrophage-mediated phagocytosis of CD11c+ DCs from BRAFV600ECD11c mice, whereas it had no effects on the viability or CCL19-dependent migration of such CD11c+ DCs or on their expression of the chemokine genes Ccl5, Ccl20, Cxcl11, and Cxcl12. Our results thus suggest that anti-SIRPα monotherapy is a promising approach to the treatment of LCH that is dependent in part on the promotion of the macrophage-mediated killing of LCH cells.


Asunto(s)
Histiocitosis de Células de Langerhans , Animales , Humanos , Ratones , Histiocitosis de Células de Langerhans/tratamiento farmacológico , Histiocitosis de Células de Langerhans/genética , Histiocitosis de Células de Langerhans/metabolismo , Bazo/metabolismo
4.
Cancer Sci ; 112(1): 16-21, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33073467

RESUMEN

The turnover of intestinal epithelial cells (IECs) is relatively rapid (3-5 days in mouse and human), and this short existence and other aspects of the homeostasis of IECs are tightly regulated by various signaling pathways including Wnt-ß-catenin signaling. Dysregulation of IEC homeostasis likely contributes to the development of intestinal inflammation and intestinal cancer. The roles of receptor protein tyrosine kinases and their downstream signaling molecules such as Src family kinases, Ras, and mTOR in homeostatic regulation of IEC turnover have recently been evaluated. These signaling pathways have been found to promote not only the proliferation of IECs but also the differentiation of progenitor cells into secretory cell types such as goblet cells. Of note, signaling by Src family kinases, Ras, and mTOR has been shown to oppose the Wnt-ß-catenin signaling pathway and thereby to limit the number of Lgr5+ intestinal stem cells or of Paneth cells. Such cross-talk of signaling pathways is important not only for proper regulation of IEC homeostasis but for the development of intestinal tumors and potentially for anticancer therapy.


Asunto(s)
Carcinogénesis/metabolismo , Células Epiteliales/metabolismo , Homeostasis/fisiología , Intestinos/patología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas ras/metabolismo , Familia-src Quinasas/metabolismo , Animales , Carcinogénesis/patología , Células Epiteliales/patología , Humanos
5.
Eur J Immunol ; 50(10): 1560-1570, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32438469

RESUMEN

Signal regulatory protein α (SIRPα) is expressed predominantly on type 2 conventional dendritic cells (cDC2s) and macrophages. We previously showed that mice systemically lacking SIRPα were resistant to experimental autoimmune encephalomyelitis (EAE). Here, we showed that deletion of SIRPα in CD11c+ cells of mice (SirpaΔDC mice) also markedly ameliorated the development of EAE. The frequency of cDCs and migratory DCs (mDCs), as well as that of Th17 cells, were significantly reduced in draining lymph nodes of SirpaΔDC mice at the onset of EAE. In addition, we found the marked reduction in the number of Th17 cells and DCs in the CNS of SirpaΔDC mice at the peak of EAE. Whereas inducible systemic ablation of SIRPα before the induction of EAE prevented disease development, that after EAE onset did not ameliorate the clinical signs of disease. We also found that EAE development was partially attenuated in mice with CD11c+ cell-specific ablation of CD47, a ligand of SIRPα. Collectively, our results suggest that SIRPα expressed on CD11c+ cells, such as cDC2s and mDCs, is indispensable for the development of EAE, being required for the priming of self-reactive Th17 cells in the periphery as well as for the inflammation in the CNS.


Asunto(s)
Sistema Nervioso Central/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Esclerosis Múltiple/inmunología , Receptores Inmunológicos/metabolismo , Células Th17/inmunología , Animales , Antígeno CD11c/metabolismo , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Inmunológicos/genética
6.
Eur J Immunol ; 49(9): 1364-1371, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31099900

RESUMEN

Nonhematopoietic stromal cells contribute to the organization and homeostasis of secondary lymphoid organs by producing cytokines and chemokines. The development and maintenance of these stromal cells are thought to be regulated by innate immune cells. Indeed, we recently showed that signal regulatory protein α (SIRPα)-positive dendritic cells (DCs) are essential for the proliferation and survival of podoplanin (Pdpn)-positive fibroblastic reticular cells (FRCs) in mouse spleen. We have now established an in vitro culture system for lymph node stromal cells (LNSCs) isolated from mouse peripheral LNs. Activated DCs and TNF-α each promoted the proliferation of cultured LNSCs, most of which were found to be Pdpn+ FRCs. Furthermore, ablation of SIRPα in CD11c+ cells attenuated this effect of DCs on LNSC proliferation. Transplantation of activated DCs together with cultured LNSCs into the renal subcapsular space markedly increased the number of ER-TR7+ stromal cells as well as induced the accumulation of T cells and increased the expression of Ccl19 in the transplants. Ablation of SIRPα in CD11c+ cells greatly impaired the development of LN-like structure in the transplants. Our findings thus suggest that SIRPα+ DCs are important for the proliferation and differentiation of Pdpn+ FRCs in peripheral LNs.


Asunto(s)
Células Dendríticas/inmunología , Fibroblastos/inmunología , Ganglios Linfáticos/inmunología , Receptores Inmunológicos/inmunología , Animales , Antígeno CD11c/inmunología , Diferenciación Celular/inmunología , Proliferación Celular/fisiología , Células Cultivadas , Homeostasis/inmunología , Inmunidad Innata/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Células del Estroma/inmunología , Linfocitos T/inmunología
7.
Proc Natl Acad Sci U S A ; 114(47): E10151-E10160, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109283

RESUMEN

In secondary lymphoid organs, development and homeostasis of stromal cells such as podoplanin (Pdpn)-positive fibroblastic reticular cells (FRCs) are regulated by hematopoietic cells, but the cellular and molecular mechanisms of such regulation have remained unclear. Here we show that ablation of either signal regulatory protein α (SIRPα), an Ig superfamily protein, or its ligand CD47 in conventional dendritic cells (cDCs) markedly reduced the number of CD4+ cDCs as well as that of Pdpn+ FRCs and T cells in the adult mouse spleen. Such ablation also impaired the survival of FRCs as well as the production by CD4+ cDCs of tumor necrosis factor receptor (TNFR) ligands, including TNF-α, which was shown to promote the proliferation and survival of Pdpn+ FRCs. CD4+ cDCs thus regulate the steady-state homeostasis of FRCs in the adult spleen via the production of TNFR ligands, with the CD47-SIRPα interaction in cDCs likely being indispensable for such regulation.


Asunto(s)
Células Dendríticas/inmunología , Fibroblastos/inmunología , Homeostasis/inmunología , Receptores Inmunológicos/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Bazo/inmunología , Animales , Antígenos CD4/genética , Antígenos CD4/inmunología , Antígeno CD47/genética , Antígeno CD47/inmunología , Supervivencia Celular , Células Dendríticas/citología , Fibroblastos/citología , Regulación de la Expresión Génica , Homeostasis/genética , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Inmunológicos/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal , Bazo/citología , Linfocitos T/citología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
8.
Cancer Sci ; 109(8): 2349-2357, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29873856

RESUMEN

Tumor cells evade immune surveillance through direct or indirect interactions with various types of immune cell, with much recent attention being focused on modifying immune cell responses as the basis for the development of new cancer treatments. Signal regulatory protein α (SIRPα) and CD47 are both transmembrane proteins that interact with each other and constitute a cell-cell communication system. SIRPα is particularly abundant in myeloid cells such as macrophages and dendritic cells, whereas CD47 is expressed ubiquitously and its expression level is elevated in cancer cells. Recent studies have shown that blockade of CD47-SIRPα interaction enhances the phagocytic activity of phagocytes such as macrophages toward tumor cells in vitro as well as resulting in the efficient eradication of tumor cells in a variety of xenograft or syngeneic mouse models of cancer. Moreover, CD47 blockade has been shown to promote the stimulation of tumor-specific cytotoxic T cells by macrophages or dendritic cells. Biological agents, such as Abs and recombinant proteins, that target human CD47 or SIRPα have been developed and are being tested in preclinical models of human cancer or in clinical trials with cancer patients. Preclinical studies have also suggested that CD47 or SIRPα blockade may have a synergistic antitumor effect in combination with immune checkpoint inhibitors that target the adaptive immune system. Targeting of the CD47-SIRPα signaling system is thus a promising strategy for cancer treatment based on modulation of both innate and acquired immune responses to tumor cells.


Asunto(s)
Antígeno CD47/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Receptores Inmunológicos/inmunología , Inmunidad Adaptativa/inmunología , Animales , Humanos , Inmunoterapia/métodos
9.
Cancer Sci ; 109(5): 1300-1308, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29473266

RESUMEN

Interaction of signal regulatory protein α (SIRPα) expressed on the surface of macrophages with its ligand CD47 expressed on target cells negatively regulates phagocytosis of the latter cells by the former. We recently showed that blocking Abs to mouse SIRPα enhanced both the Ab-dependent cellular phagocytosis (ADCP) activity of mouse macrophages for Burkitt's lymphoma Raji cells opsonized with an Ab to CD20 (rituximab) in vitro as well as the inhibitory effect of rituximab on the growth of tumors formed by Raji cells in nonobese diabetic (NOD)/SCID mice. However, the effects of blocking Abs to human SIRPα in preclinical cancer models have remained unclear given that such Abs have failed to interact with endogenous SIRPα expressed on macrophages of immunodeficient mice. With the use of Rag2-/- γc-/- mice harboring a transgene for human SIRPα under the control of human regulatory elements (hSIRPα-DKO mice), we here show that a blocking Ab to human SIRPα significantly enhanced the ADCP activity of macrophages derived from these mice for human cancer cells. The anti-human SIRPα Ab also markedly enhanced the inhibitory effect of rituximab on the growth of tumors formed by Raji cells in hSIRPα-DKO mice. Our results thus suggest that the combination of Abs to human SIRPα with therapeutic Abs specific for tumor antigens warrants further investigation for potential application to cancer immunotherapy. In addition, humanized mice, such as hSIRPα-DKO mice, should prove useful for validation of the antitumor effects of checkpoint inhibitors before testing in clinical trials.


Asunto(s)
Antígenos de Diferenciación/inmunología , Antineoplásicos Inmunológicos/administración & dosificación , Linfoma de Burkitt/tratamiento farmacológico , Receptores Inmunológicos/inmunología , Rituximab/administración & dosificación , Animales , Antígenos de Diferenciación/genética , Antineoplásicos Inmunológicos/farmacología , Linfoma de Burkitt/genética , Linfoma de Burkitt/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunoterapia , Macrófagos/citología , Macrófagos/inmunología , Ratones , Fagocitosis , Receptores Inmunológicos/genética , Rituximab/farmacología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Biochem Biophys Res Commun ; 504(1): 109-114, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30173891

RESUMEN

Intestinal epithelial cells (IECs) play a pivotal role in the maintenance of the integrity and barrier function of the intestinal epithelium. Dysfunctions of IECs are thought to participate in the disruption of the intestinal epithelial barrier, resulting in gastrointestinal diseases, such as colitis and colorectal cancer. Here we show that IEC-specific COOH-terminal Src kinase (Csk)-deficient mice (Csk CKO mice) manifested the increased susceptibility to dextran sodium sulfate (DSS)-induced colitis, a model of inflammatory bowel disease. DSS-treated Csk CKO mice also exhibited the significantly elevated intestinal permeability. Following DSS treatment, Csk CKO mice exhibited the higher proliferative activity of colonic epithelial cells and the increased number of apoptotic cells in the colon compared with that apparent for control mice. Moreover, the abundance of the tight junction protein occludin, which regulates cell-cell adhesion as well as epithelial permeability, was markedly reduced in the colon of DSS-treated Csk CKO mice. These results thus suggest that Csk in IECs plays important roles in the regulation of the intestinal epithelial barrier function and protection against colitis.


Asunto(s)
Colitis/metabolismo , Mucosa Intestinal/metabolismo , Familia-src Quinasas/fisiología , Uniones Adherentes/metabolismo , Animales , Apoptosis , Proteína Tirosina Quinasa CSK , Proliferación Celular , Colitis/inducido químicamente , Colitis/patología , Colon/metabolismo , Sulfato de Dextran , Mucosa Intestinal/citología , Masculino , Ratones , Ratones Noqueados , Permeabilidad , Proteínas de Uniones Estrechas/metabolismo , Familia-src Quinasas/genética
11.
Biochem Biophys Res Commun ; 498(4): 824-829, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29530528

RESUMEN

In the mouse olfactory bulb (OB), interneurons such as granule cells and periglomerular cells are continuously replaced by adult-born neurons, which are generated in the subventricular zone (SVZ) of the brain. We have now investigated the role of commensal bacteria in regulation of such neuronal cell turnover in the adult mouse brain. Administration of mixture of antibiotics to specific pathogen-free (SPF) mice markedly attenuated the incorporation of bromodeoxyuridine (BrdU) into the SVZ cells. The treatment with antibiotics also reduced newly generated BrdU-positive neurons in the mouse OB. In addition, the incorporation of BrdU into the SVZ cells of germ-free (GF) mice was markedly reduced compared to that apparent for SPF mice. In contrast, the reduced incorporation of BrdU into the SVZ cells of GF mice was recovered by their co-housing with SPF mice, suggesting that commensal bacteria promote the incorporation of BrdU into the SVZ cells. Finally, we found that administration of ampicillin markedly attenuated the incorporation of BrdU into the SVZ cells of SPF mice. Our results thus suggest that ampicillin-sensitive commensal bacteria regulate the neurogenesis in the SVZ of adult mouse brain.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Ventrículos Laterales/crecimiento & desarrollo , Ventrículos Laterales/microbiología , Neurogénesis , Bulbo Olfatorio/crecimiento & desarrollo , Bulbo Olfatorio/microbiología , Simbiosis , Ampicilina/farmacología , Animales , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Interneuronas/citología , Interneuronas/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/microbiología
12.
Proc Natl Acad Sci U S A ; 112(31): E4264-71, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195794

RESUMEN

Intestinal epithelial cells contribute to regulation of intestinal immunity in mammals, but the detailed molecular mechanisms of such regulation have remained largely unknown. Stomach-cancer-associated protein tyrosine phosphatase 1 (SAP-1, also known as PTPRH) is a receptor-type protein tyrosine phosphatase that is localized specifically at microvilli of the brush border in gastrointestinal epithelial cells. Here we show that SAP-1 ablation in interleukin (IL)-10-deficient mice, a model of inflammatory bowel disease, resulted in a marked increase in the severity of colitis in association with up-regulation of mRNAs for various cytokines and chemokines in the colon. Tyrosine phosphorylation of carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 20, an intestinal microvillus-specific transmembrane protein of the Ig superfamily, was greatly increased in the intestinal epithelium of the SAP-1-deficient animals, suggesting that this protein is a substrate for SAP-1. Tyrosine phosphorylation of CEACAM20 by the protein tyrosine kinase c-Src and the consequent association of CEACAM20 with spleen tyrosine kinase (Syk) promoted the production of IL-8 in cultured cells through the activation of nuclear factor-κB (NF-κB). In addition, SAP-1 and CEACAM20 were found to form a complex through interaction of their ectodomains. SAP-1 and CEACAM20 thus constitute a regulatory system through which the intestinal epithelium contributes to intestinal immunity.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Colitis/enzimología , Colitis/prevención & control , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Animales , Recuento de Células , Quimiocinas/genética , Quimiocinas/metabolismo , Colitis/patología , Colon/patología , Femenino , Células Caliciformes/metabolismo , Células Caliciformes/patología , Células HEK293 , Humanos , Interleucina-10/deficiencia , Interleucina-10/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica , Transporte de Proteínas , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/deficiencia , Quinasa Syk , Dominios Homologos src , Familia-src Quinasas/metabolismo
13.
Genes Cells ; 20(7): 578-89, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25908210

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 1 and CEACAM20, immunoglobulin superfamily members, are predominantly expressed in intestinal epithelial cells (IECs) and co-localized at the apical surface of these cells. We here showed that the expression of mouse CEACAM1 and CEACAM20 at both mRNA and protein levels was markedly reduced in IECs of the small intestine by the treatment of mice with antibiotics against Gram-positive bacteria. The expression of both proteins was also decreased in IECs of the small intestine from germ-free mice, compared with that from control specific-pathogen-free mice. Exposure of intestinal organoids to IFN-γ markedly increased the expression of either CEACAM1 or CEACAM20, whereas the exposure to TNF-α increased the expression of the former protein, but not that of the latter. In contrast, the expression of CEACAM20, but not of CEACAM1, in intestinal organoids was markedly increased by exposure to butyrate, a short-chain fatty acid produced by bacterial fermentation in the intestine. Collectively, our results suggest that Gram-positive bacteria promote the mRNA expression of CEACAM1 or CEACAM20 in the small intestine. Inflammatory cytokines or butyrate likely participates in such effects of commensal bacteria.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Regulación de la Expresión Génica , Bacterias Grampositivas/metabolismo , Intestino Delgado/metabolismo , ARN Mensajero/metabolismo , Animales , Antibacterianos/farmacología , Butiratos/metabolismo , Antígeno Carcinoembrionario/genética , Moléculas de Adhesión Celular/genética , Células Epiteliales/metabolismo , Ácidos Grasos Volátiles/metabolismo , Bacterias Grampositivas/efectos de los fármacos , Interferón gamma/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/citología , Intestino Delgado/microbiología , Intestinos/citología , Intestinos/microbiología , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
14.
Genes Cells ; 20(6): 451-63, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25818708

RESUMEN

Signal regulatory protein α (SIRPα), an immunoglobulin superfamily protein that is expressed predominantly in myeloid lineage cells such as dendritic cells (DCs) or macrophages, mediates cell-cell signaling. In the immune system, SIRPα is thought to be important for homeostasis of DCs, but it remains unclear whether SIRPα intrinsic to DCs is indeed indispensable for such functional role. Thus, we here generated the mice, in which SIRPα was specifically ablated in CD11c(+) DCs (Sirpa(Δ) (DC) ). Sirpa(Δ) (DC) mice manifested a marked reduction of CD4(+) CD8α(-) conventional DCs (cDCs) in the secondary lymphoid organs, as well as of Langerhans cells in the epidermis. Such reduction of cDCs in Sirpa(Δ) (DC) mice was comparable to that apparent with the mice, in which SIRPα was systemically ablated. Expression of SIRPα in DCs was well correlated with that of either endothelial cell-selective adhesion molecule (ESAM) or Epstein-Barr virus-induced molecule 2 (EBI2), both of which were also implicated in the regulation of DC homeostasis. Indeed, ESAM(+) or EBI2(+) cDCs were markedly reduced in the spleen of Sirpa(Δ) (DC) mice. Thus, our results suggest that SIRPα intrinsic to CD11c(+) DCs is essential for homeostasis of cDCs in the secondary lymphoid organs and skin.


Asunto(s)
Células Dendríticas/metabolismo , Homeostasis , Tejido Linfoide/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Antígenos CD8/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Epidérmicas , Ganglios Linfáticos/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/metabolismo , Bazo/citología
15.
J Biol Chem ; 289(10): 6451-6461, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24451369

RESUMEN

Vascular endothelial cells (ECs) are continuously exposed to shear stress (SS) generated by blood flow. Such stress plays a key role in regulation of various aspects of EC function including cell proliferation and motility as well as changes in cell morphology. Vascular endothelial-protein-tyrosine phosphatase (VE-PTP) is an R3-subtype PTP that possesses multiple fibronectin type III-like domains in its extracellular region and is expressed specifically in ECs. The role of VE-PTP in EC responses to SS has remained unknown, however. Here we show that VE-PTP is diffusely localized in ECs maintained under static culture conditions, whereas it undergoes rapid accumulation at the downstream edge of the cells relative to the direction of flow in response to SS. This redistribution of VE-PTP triggered by SS was found to require its extracellular and transmembrane regions and was promoted by integrin engagement of extracellular matrix ligands. Inhibition of actin polymerization or of Cdc42, Rab5, or Arf6 activities attenuated the SS-induced redistribution of VE-PTP. VE-PTP also underwent endocytosis in the static and SS conditions. SS induced the polarized distribution of internalized VE-PTP. Such an effect was promoted by integrin engagement of fibronectin but prevented by inhibition of Cdc42 activity or of actin polymerization. In addition, depletion of VE-PTP by RNA interference in human umbilical vein ECs blocked cell elongation in the direction of flow induced by SS. Our results suggest that the polarized redistribution of VE-PTP in response to SS plays an important role in the regulation of EC function by blood flow.


Asunto(s)
Aumento de la Célula , Endotelio Vascular/fisiología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Resistencia al Corte , Estrés Mecánico , Actinas/metabolismo , Animales , Circulación Sanguínea , Línea Celular Tumoral , Citoesqueleto/metabolismo , Citoesqueleto/fisiología , Endotelio Vascular/enzimología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones , Interferencia de ARN , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Proteína de Unión al GTP cdc42/metabolismo
16.
Methods ; 65(2): 254-9, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24091004

RESUMEN

Signal regulatory protein α (SIRPα), also known as SHPS-1/SIRPA, is an immunoglobulin superfamily protein that binds to the protein tyrosine phosphatases Shp1 and Shp2 through its cytoplasmic region and is predominantly expressed in dendritic cells and macrophages. CD47, a widely expressed transmembrane protein, is a ligand for SIRPα, with the two proteins constituting a cell-cell communication system. It was previously demonstrated that the CD47-SIRPα signaling pathway is important for prevention of clearance by splenic macrophages of red blood cells or platelets from the bloodstream. In addition, this signaling pathway is also implicated in homeostatic regulation of dendritic cells and development of autoimmunity. Here we describe the detailed protocols for methods that were used in our recent studies to study the role of the CD47-SIRPα signaling pathway in autoimmunity. We also demonstrate that hematopoietic SIRPα as well as nonhematopoietic CD47 are important for development of experimental autoimmune encephalomyelitis. Thus, we here strengthen the importance of experimental animal models as well as other methods for the study of molecular pathogenesis of autoimmunity.


Asunto(s)
Antígeno CD47/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Receptores Inmunológicos/metabolismo , Transducción de Señal/fisiología , Animales , Antígeno CD47/genética , Encefalomielitis Autoinmune Experimental/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Inmunológicos/genética
17.
J Immunol ; 188(11): 5397-407, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22539788

RESUMEN

Dendritic cells (DCs) promote immune responses to foreign Ags and immune tolerance to self-Ags. Deregulation of DCs is implicated in autoimmunity, but the molecules that regulate DCs to protect against autoimmunity have remained unknown. In this study, we show that mice lacking the protein tyrosine phosphatase Shp1 specifically in DCs develop splenomegaly associated with more CD11c(+) DCs. Splenic DCs from the mutant mice showed upregulation of CD86 and CCR7 expression and of LPS-induced production of proinflammatory cytokines. The mice manifested more splenic Th1 cells, consistent with the increased ability of their DCs to induce production of IFN-γ by Ag-specific T cells in vitro. The number of splenic CD5(+)CD19(+) B-1a cells and the serum concentrations of Igs M and G2a were also increased in the mutant mice. Moreover, aged mutant mice developed glomerulonephritis and interstitial pneumonitis together with increased serum concentrations of autoantibodies. Shp1 is thus a key regulator of DC functions that protects against autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/genética , Diferenciación Celular/inmunología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Células TH1/inmunología , Animales , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Antígeno CD11c/biosíntesis , Diferenciación Celular/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Esplenomegalia/genética , Esplenomegalia/inmunología , Esplenomegalia/patología , Células TH1/citología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
18.
Regen Med ; 19(3): 119-133, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38449425

RESUMEN

Aim: This study aimed to evaluate the trends in organoid culture research within the field of regenerative medicine from 2002 to 2022. Methods: The worldwide distribution of organoid research in regenerative medicine articles indexed in the Scopus database was analyzed. Result: A total of 840 documents were analyzed, averaging 42 publications annually. The USA (n = 296) led in publications, followed by China (n = 127), Japan (n = 91) and the UK (n = 75). Since 2011, research has surged, particularly in China, which emerged as a prominent center. Conclusion: The findings highlight significant growth in organoid research, promising future organ transplantation. Research trends integrate tissue engineering, gene modification and induced pluripotent stem cell technologies, reflecting a move toward personalized medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas , Medicina Regenerativa , Organoides , Ingeniería de Tejidos , Bibliometría
19.
J Immunol ; 187(1): 291-7, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21632712

RESUMEN

The molecular basis for formation of lymphoid follicle and its homeostasis in the secondary lymphoid organs remains unclear. Signal regulatory protein α (SIRPα), an Ig superfamily protein that is predominantly expressed in dendritic cells or macrophages, mediates cell-cell signaling by interacting with CD47, another Ig superfamily protein. In this study, we show that the size of the T cell zone as well as the number of CD4(+) T cells were markedly reduced in the spleen of mice bearing a mutant (MT) SIRPα that lacks the cytoplasmic region compared with those of wild-type mice. In addition, the expression of CCL19 and CCL21, as well as of IL-7, which are thought to be important for development or homeostasis of the T cell zone, was markedly decreased in the spleen of SIRPα MT mice. By the use of bone marrow chimera, we found that hematopoietic SIRPα is important for development of the T cell zone as well as the expression of CCL19 and CCL21 in the spleen. The expression of lymphotoxin and its receptor, lymphotoxin ß receptor, as well as the in vivo response to lymphotoxin ß receptor stimulation were also decreased in the spleen of SIRPα MT mice. CD47-deficient mice also manifested phenotypes similar to SIRPα MT mice. These data suggest that SIRPα as well as its ligand CD47 are thus essential for steady-state homeostasis of T cells in the spleen.


Asunto(s)
Homeostasis/inmunología , Receptores Inmunológicos/fisiología , Bazo/citología , Bazo/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Recuento de Linfocito CD4 , Antígeno CD47/genética , Antígeno CD47/metabolismo , Antígeno CD47/fisiología , Tamaño de la Célula , Homeostasis/genética , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Inmunológicos/genética , Bazo/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo
20.
J Gastroenterol ; 58(5): 444-457, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36739585

RESUMEN

BACKGROUND: Amino acid transporters play an important role in supplying nutrition to cells and are associated with cell proliferation. L-type amino acid transporter 1 (LAT1) is highly expressed in many types of cancers and promotes tumor growth; however, how LAT1 affects tumor development is not fully understood. METHODS: To investigate the role of LAT1 in intestinal tumorigenesis, mice carrying LAT1 floxed alleles that also expressed Cre recombinase from the promoter of gene encoding Villin were crossed to an ApcMin/+ background (LAT1fl/fl; vil-cre; ApcMin/+), which were subject to analysis; organoids derived from those mice were also analyzed. RESULTS: This study showed that LAT1 was constitutively expressed in normal crypt base cells, and its conditional deletion in the intestinal epithelium resulted in fewer Paneth cells. LAT1 deletion reduced tumor size and number in the small intestine of ApcMin/+ mice. Organoids derived from LAT1-deleted ApcMin/+ intestinal crypts displayed fewer spherical organoids with reduced Wnt/ß-catenin target gene expression, suggesting a low tumor-initiation capacity. Wnt3 expression was decreased in the absence of LAT1 in the intestinal epithelium, suggesting that loss of Paneth cells due to LAT1 deficiency reduced the risk of tumor initiation by decreasing Wnt3 production. CONCLUSIONS: LAT1 affects intestinal tumor development in a cell-extrinsic manner through reduced Wnt3 expression in Paneth cells. Our findings may partly explain how nutrient availability can affect the risk of tumor development in the intestines.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon , Sistema de Transporte de Aminoácidos y+L , Neoplasias Intestinales , Células de Paneth , Animales , Ratones , Transformación Celular Neoplásica/genética , Mucosa Intestinal/patología , Neoplasias Intestinales/metabolismo , Intestino Delgado/patología , Intestinos , Células de Paneth/metabolismo , Células de Paneth/patología , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Sistema de Transporte de Aminoácidos y+L/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA