Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
STAR Protoc ; 4(3): 102472, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37515759

RESUMEN

Senescence-associated (SA) CD4+ T cells, which increase with age, may underlie the development of autoimmunity and chronic inflammation, but their pathological function remains understudied. Here, we present a protocol to isolate CD153+ SA-T cells and evaluate their characteristic responses upon T cell receptor stimulation. We describe steps for the isolation of CD153+ SA-T cells using flow cytometry and in vitro culture with stimulatory antibodies against CD3, CD28, and CD153. We then detail the assessment of the proliferation capacity and cytokine production. For complete details on the use and execution of this protocol, please refer to Fukushima et al. (2022).1.


Asunto(s)
Anticuerpos , Linfocitos T , Animales , Ratones , Citometría de Flujo , Bioensayo , Linfocitos T CD4-Positivos
2.
Sci Immunol ; 7(76): eabj8760, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36269840

RESUMEN

Invariant natural killer T (iNKT) cells are a group of innate-like T lymphocytes that recognize lipid antigens. They are supposed to be tissue resident and important for systemic and local immune regulation. To investigate the heterogeneity of iNKT cells, we recharacterized iNKT cells in the thymus and peripheral tissues. iNKT cells in the thymus were divided into three subpopulations by the expression of the natural killer cell receptor CD244 and the chemokine receptor CXCR6 and designated as C0 (CD244-CXCR6-), C1 (CD244-CXCR6+), or C2 (CD244+CXCR6+) iNKT cells. The development and maturation of C2 iNKT cells from C0 iNKT cells strictly depended on IL-15 produced by thymic epithelial cells. C2 iNKT cells expressed high levels of IFN-γ and granzymes and exhibited more NK cell-like features, whereas C1 iNKT cells showed more T cell-like characteristics. C2 iNKT cells were influenced by the microbiome and aging and suppressed the expression of the autoimmune regulator AIRE in the thymus. In peripheral tissues, C2 iNKT cells were circulating that were distinct from conventional tissue-resident C1 iNKT cells. Functionally, C2 iNKT cells protected mice from the tumor metastasis of melanoma cells by enhancing antitumor immunity and promoted antiviral immune responses against influenza virus infection. Furthermore, we identified human CD244+CXCR6+ iNKT cells with high cytotoxic properties as a counterpart of mouse C2 iNKT cells. Thus, this study reveals a circulating subset of iNKT cells with NK cell-like properties distinct from conventional tissue-resident iNKT cells.


Asunto(s)
Células T Asesinas Naturales , Ratones , Humanos , Animales , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología , Interleucina-15 , Antivirales , Granzimas , Receptores de Células Asesinas Naturales , Receptores de Quimiocina/metabolismo , Lípidos
3.
Cell Rep ; 40(12): 111373, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36130493

RESUMEN

With age, senescence-associated (SA) CD4+ T cells that are refractory to T cell receptor (TCR) stimulation are increased along with spontaneous germinal center (Spt-GC) development prone to autoantibody production. We demonstrate that CD153 and its receptor CD30 are expressed in SA-T and Spt-GC B cells, respectively, and deficiency of either CD153 or CD30 results in the compromised increase of both cell types. CD153 engagement on SA-T cells upon TCR stimulation causes association of CD153 with the TCR/CD3 complex and restores TCR signaling, whereas CD30 engagement on GC B cells induces their expansion. Administration of an anti-CD153 antibody blocking the interaction with CD30 suppresses the increase in both SA-T and Spt-GC B cells with age and ameliorates lupus in lupus-prone mice. These results suggest that the molecular interaction of CD153 and CD30 plays a central role in the reciprocal activation of SA-T and Spt-GC B cells, leading to immunosenescent phenotypes and autoimmunity.


Asunto(s)
Ligando CD30/metabolismo , Receptores de Antígenos de Linfocitos T , Linfocitos T , Animales , Complejo CD3/metabolismo , Centro Germinal , Antígeno Ki-1/metabolismo , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
4.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34813503

RESUMEN

Tertiary lymphoid tissues (TLTs) facilitate local T and B cell interactions in chronically inflamed organs. However, the cells and molecular pathways that govern TLT formation are poorly defined. Here, we identified TNF superfamily CD153/CD30 signaling between 2 unique age-dependent lymphocyte subpopulations, CD153+PD-1+CD4+ senescence-associated T (SAT) cells and CD30+T-bet+ age-associated B cells (ABCs), as a driver for TLT expansion. SAT cells, which produced ABC-inducing factors IL-21 and IFN-γ, and ABCs progressively accumulated within TLTs in aged kidneys after injury. Notably, in kidney injury models, CD153 or CD30 deficiency impaired functional SAT cell induction, which resulted in reduced ABC numbers and attenuated TLT formation with improved inflammation, fibrosis, and renal function. Attenuated TLT formation after transplantation of CD153-deficient bone marrow further supported the importance of CD153 in immune cells. Clonal analysis revealed that SAT cells and ABCs in the kidneys arose from both local differentiation and recruitment from the spleen. In the synovium of aged rheumatoid arthritis patients, T peripheral helper/T follicular helper cells and ABCs also expressed CD153 and CD30, respectively. Together, our data reveal a previously unappreciated function of CD153/CD30 signaling in TLT formation and propose targeting the CD153/CD30 signaling pathway as a therapeutic target for slowing kidney disease progression.


Asunto(s)
Lesión Renal Aguda/inmunología , Envejecimiento/inmunología , Ligando CD30/inmunología , Antígeno Ki-1/inmunología , Tejido Linfoide/inmunología , Transducción de Señal/inmunología , Lesión Renal Aguda/genética , Envejecimiento/genética , Animales , Ligando CD30/genética , Linfocitos T CD4-Positivos/inmunología , Antígeno Ki-1/genética , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/genética
5.
Int J Biol Macromol ; 180: 718-728, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33753200

RESUMEN

Signal-induced proliferation-associated protein 1 (SIPA1) is highly expressed and mainly located in the nucleus in some breast cancer cell lines and clinical tumor tissues. Previous study revealed that nuclear localization of SIPA1 is functionally involved in breast cancer metastasis in the lymphatic gland. In the current study, we identified a non-typical region (140-179aa) of SIPA1 as a novel nuclear localization region (NLR) which is crucial for translocating the proteins into the nucleus in HEK293 cells and breast cancer cells. This region contained one basic amino acid, His160, and had no common features of typical nuclear localization signals. In addition, overexpressing SIPA1 without NLR could suppress breast cancer cell proliferation but could not promote cell migration in MCF7 cells. Furthermore, we found that a high expression of SIPA1 upregulated the expression of ABCB1, encoding multi-drug resistance protein MDR1, and promoted the resistance to epirubicin in breast cancer cells, while this effect was largely abolished in the cells with the expression of NLR-deleted SIPA1. This study overall, identified a nuclear localization-dependent region determining the nuclear distribution of SIPA1 and its regulation on epirubicin-sensitivity in breast cancer cells, which could be a potential drug target to facilitate the development of breast cancer chemotherapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Núcleo Celular/metabolismo , Epirrubicina/farmacología , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Aminoácidos/genética , Aminoácidos/metabolismo , Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/genética , Células HEK293 , Humanos , Células MCF-7 , Microscopía Fluorescente , Mutación , Señales de Localización Nuclear/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética
6.
Int Immunol ; 33(3): 171-182, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33038259

RESUMEN

T-cell development depends on the thymic microenvironment, in which endothelial cells (ECs) play a vital role. Interestingly, vascular permeability of the thymic cortex is lower than in other organs, suggesting the existence of a blood-thymus barrier (BTB). On the other hand, blood-borne molecules and dendritic cells bearing self-antigens are accessible to the medulla, facilitating central tolerance induction, and continuous T-precursor immigration and mature thymocyte egress occur through the vessels at the cortico-medullary junction (CMJ). We found that claudin-5 (Cld5), a membrane protein of tight junctions, was expressed in essentially all ECs of the cortical vasculatures, whereas approximately half of the ECs of the medulla and CMJ lacked Cld5 expression. An intravenously (i.v.) injected biotin tracer hardly penetrated cortical Cld5+ vessels, but it leaked into the medullary parenchyma through Cld5- vessels. Cld5 expression in an EC cell line caused a remarkable increase in trans-endothelial resistance in vitro, and the biotin tracer leaked from the cortical vasculatures in Cldn5-/- mice. Furthermore, i.v.-injected sphingosine-1 phosphate distributed selectively into the medulla through the Cld5- vessels, probably ensuring the egress of CD3high mature thymocytes from Cld5- vessels at the CMJ. These results suggest that distinct Cld5 expression profiles in the cortex and medulla may control the BTB and the T-cell gateway to blood circulation, respectively.


Asunto(s)
Permeabilidad Capilar/fisiología , Claudina-5/metabolismo , Linfocitos T/metabolismo , Timo/metabolismo , Uniones Estrechas/fisiología , Animales , Diferenciación Celular/inmunología , Línea Celular , Claudina-5/biosíntesis , Células Endoteliales/metabolismo , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Linfocitos T/citología , Timocitos/metabolismo
7.
Stem Cell Res ; 49: 102099, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33296812

RESUMEN

SIPA1, a GTPase activating protein that negatively regulates Ras-related protein (Rap), is a potential modulator of tumor metastasis and recurrence. In this study, we first showed that SIPA1 facilitated the stemness features of breast cancer cells, such as of tumorsphere formation capability and the expression of stemness marker CD44. In addition, SIPA1 promoted the expression of four stemness-associated transcription factors through increasing the expression of SMAD2 and SMAD3 in vitro and in vivo. The stemness features were abolished by blocking the phosphorylation of SMAD3 with its specific inhibitor SIS3. Furthermore, SIPA1 decreased the breast cancer cell sensitivity to chemotherapy drugs. This effect was, however, competitively reversed by blocking the SMAD3 phosphorylation by SIS3 treatment in breast cancer cells. Taken together, SIPA1 promotes and sustains the stemness of breast cancer cells and their resistance to chemotherapy by increasing the expression of SMAD2 and SMAD3, and blocking SMAD3 phosphorylation could suppress the cancer cell stemness and increase the sensitivity to chemotherapy in breast cancer cells expressing a high level of SIPA1.


Asunto(s)
Neoplasias de la Mama , Proteínas Activadoras de GTPasa/genética , Células Madre Neoplásicas/citología , Proteínas Nucleares/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Recurrencia Local de Neoplasia , Fosforilación , Proteína Smad2/genética , Proteína smad3
8.
J Immunol ; 205(8): 2008-2015, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32907997

RESUMEN

Immune complexes (ICs) in blood are efficiently removed mainly by liver reticuloendothelial systems consisting of sinusoidal endothelial cells and Kupffer cells expressing FcγR. The bone marrow (BM) also has sinusoidal vasculatures, and sinusoidal BM endothelial cells (BMECs) bear unique function, including hematopoietic niches and traffic regulation of hematopoietic cells. In this study, we found that sinusoidal BMECs express FcγRIIb2, which is markedly increased in anemic conditions or by the administration of erythropoietin (Epo) in healthy mice. BMECs expressed Epo receptor (EpoR), and the Epo-induced increase in FcγRIIb2 expression was abolished in Epor-/- ::HG1-Epor transgenic mice, which lack EpoR in BMECs except for BM erythroblasts, suggesting the effect was directly mediated via EpoR on BMECs. Further, although BMECs hardly captured i.v.-injected soluble ICs in healthy mice, Epo administration induced a remarkable increase in the uptake of ICs in a FcγRIIb-dependent manner. Enhancement of the IC incorporation capacity by Epo was also observed in cultured BMECs in vitro, suggesting the direct effect of Epo on BMECs. Moreover, we found that i.v.-injected ICs in Epo-treated mice were more rapidly removed from the circulation than in PBS-treated mice. These results reveal a novel function of BMECs to efficiently remove circulating blood-borne ICs in an FcγRIIb2-mediated manner.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Células de la Médula Ósea/inmunología , Células Endoteliales/inmunología , Eritropoyetina/inmunología , Receptores de IgG/inmunología , Animales , Complejo Antígeno-Anticuerpo/sangre , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Eritropoyetina/sangre , Eritropoyetina/genética , Ratones , Ratones Noqueados , Receptores de IgG/sangre , Receptores de IgG/genética
9.
Int J Clin Oncol ; 25(5): 787-789, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32180033
10.
Int Immunol ; 32(4): 223-231, 2020 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-31967307

RESUMEN

Acquired immune function shows recognizable changes over time with organismal aging. These changes include T-cell dysfunction, which may underlie diminished resistance to infection and possibly various chronic age-associated diseases in the elderly. T-cell dysfunction may occur at distinct stages, from naive cells to the end stages of differentiation during immune responses. The thymus, which generates naive T cells, shows unusually early involution resulting in progressive reduction of T-cell output after adolescence, but peripheral T-cell numbers are maintained through antigen-independent homeostatic proliferation of naive T cells driven by the major histocompatibility complex associated with self-peptides and homeostatic cytokines, retaining the diverse repertoire. However, extensive homeostatic proliferation may lead to the emergence of dysfunctional CD4+ T cells with features resembling senescent cells, termed senescence-associated T (SA-T) cells, which increase and accumulate with age. In situations such as chronic viral infection, T-cell dysfunction may also develop via persistent antigen stimulation, termed exhaustion, preventing possible immunopathology due to excessive immune responses. Exhausted T cells are developed through the effects of checkpoint receptors such as PD-1 and may be reversed with the receptor blockade. Of note, although defective in their regular T-cell antigen-receptor-mediated proliferation, SA-T cells secrete abundant pro-inflammatory factors such as osteopontin, reminiscent of an SA-secretory phenotype. A series of experiments in mouse models indicated that SA-T cells are involved in systemic autoimmunity as well as chronic tissue inflammation following tissue stresses. In this review, we discuss the physiological aspects of T-cell dysfunction associated with aging and its potential pathological involvement in age-associated diseases and possibly cancer.


Asunto(s)
Senescencia Celular/inmunología , Linfocitos T/inmunología , Animales , Humanos , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/patología
11.
Sci Rep ; 9(1): 10144, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31300681

RESUMEN

Current clinically approved biomarkers for the PD-1 blockade cancer immunotherapy are based entirely on the properties of tumour cells. With increasing awareness of clinical responses, more precise biomarkers for the efficacy are required based on immune properties. In particular, expression levels of immune checkpoint-associated molecules such as PD-1, PD-L1, and CTLA-4 would be critical to evaluate the immune state of individuals. Although quantification of their soluble form leased from the membrane will provide quick evaluation of patients' immune status, available methods such as enzyme-linked immunosorbent assays to measure these soluble factors have limitations in sensitivity and reproducibility for clinical use. To overcome these problems, we developed a rapid and sensitive immunoassay system based on chemiluminescent magnetic technology. The system is fully automated, providing high reproducibility. Application of this system to plasma of patients with several types of tumours demonstrated that soluble PD-1, PD-L1, and CTLA-4 levels were increased compared to those of healthy controls and varied among tumour types. The sensitivity and detection range were sufficient for evaluating plasma concentrations before and after the surgical ablation of cancers. Therefore, our newly developed system shows potential for accurate detection of soluble PD-1, PD-L1, and CTLA-4 levels in the clinical practice.


Asunto(s)
Antígeno B7-H1/sangre , Biomarcadores de Tumor/sangre , Antígeno CTLA-4/sangre , Inmunoensayo/métodos , Receptor de Muerte Celular Programada 1/sangre , Automatización de Laboratorios , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Células Renales/sangre , Estudios de Casos y Controles , Femenino , Humanos , Neoplasias Renales/sangre , Luminiscencia , Neoplasias Pulmonares/sangre , Mieloma Múltiple/sangre , Neoplasias Ováricas/sangre , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
PLoS One ; 14(7): e0215883, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31291255

RESUMEN

Innate CD8αα+ cells, also referred to as iCD8α cells, are TCR-negative intraepithelial lymphocytes (IEL) possessing cytokine and chemokine profiles and functions related to innate immune cells. iCD8α cells constitute an important source of osteopontin in the intestinal epithelium. Osteopontin is a pleiotropic cytokine with diverse roles in bone and tissue remodeling, but also has relevant functions in the homeostasis of immune cells. In this report, we present evidence for the role of iCD8α cells in the homeostasis of TCR-negative NKp46+NK1.1+ IEL (ILC1-like). We also show that the effect of iCD8α cells on ILC1-like IEL is enhanced in vitro by osteopontin. We show that in the absence of iCD8α cells, the number of NKp46+NK1.1+ IEL is significantly reduced. These ILC1-like cells are involved in intestinal pathogenesis in the anti-CD40 mouse model of intestinal inflammation. Reduced iCD8α cell numbers results in a milder form of intestinal inflammation in this disease model, whereas treatment with osteopontin increases disease severity. Collectively, our results suggest that iCD8α cells promote survival of NKp46+NK1.1+ IEL, which significantly impacts the development of intestinal inflammation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Mucosa Intestinal/inmunología , Linfocitos Intraepiteliales/inmunología , Animales , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Supervivencia Celular/inmunología , Gastroenteritis/etiología , Gastroenteritis/inmunología , Gastroenteritis/patología , Homeostasis/inmunología , Inmunidad Innata , Mucosa Intestinal/patología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Osteopontina/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
13.
J Immunol ; 203(1): 167-177, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31085589

RESUMEN

Mainstream CD8+ and CD4+ T cells of αß lineage are developed in the thymus through TCR-mediated selection in the context of MHC class I and MHC class II in association with self-peptides, respectively. In addition, minor αßT cells bearing invariant TCRs, NKT cells, and mucosal-associated invariant T cells are selected via MHC-like molecules, CD1d, and MR1 complexed with nonpeptide Ags, respectively, parts of which express neither CD4 nor CD8. In this study, we indicate that bone marrow (BM), but barely other lymphoid tissues, harbors CD4/CD8 double-negative αßT cells with an apparently diverse TCR repertoire at considerable proportions in healthy adult mice. The BM-resident double-negative αßT (BMDNT) cells are developed in the thymus in a Notch and IL-7-dependent manner but independently of known restriction elements, including MHC class I, MHC class II, CD1d, and MR1. These cells are sustained in BM throughout the adult stage with "homeostatic" proliferation via IL-1ß derived from normal myeloid cells dominating the BM environment. Although BMDNT cells secrete a unique set of cytokines, including IL-17, GM-CSF, IL-3, and CCL chemokines on TCR stimulation, these T cells also express a series of NK receptors and exhibit a potent NK-like cytotoxic activity. Furthermore, BMDNT cells show robustly accelerated proliferation and activation following systemic administration of TLR ligands likely through the enhanced production of IL-1ß by myeloid cells in situ. Our results suggest that αßT lineage cells that are developed in the thymus by default of TCR-mediated selection are maintained and differentiated to innate-like T cells in BM and may play a role in innate immunity in the hematopoietic environment.


Asunto(s)
Médula Ósea/fisiología , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/inmunología , Timo/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Homeostasis , Inmunidad Innata , Interleucina-1beta/metabolismo , Interleucina-7/genética , Interleucina-7/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Cancer Sci ; 110(4): 1317-1330, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30767320

RESUMEN

Adult long-term hematopoiesis depends on sustaining hematopoietic stem/progenitor cells (HSPC) in bone marrow (BM) niches, where their balance of quiescence, self-renewal, and hematopoietic differentiation is tightly regulated. Although various BM stroma cells that produce niche factors have been identified, regulation of the intrinsic responsiveness of HSPC to the niche factors remains elusive. We previously reported that mice deficient for Sipa1, a Rap1 GTPase-activating protein, develop diverse hematopoietic disorders of late onset. Here we showed that transplantation of BM cells expressing membrane-targeted C3G (C3G-F), a Rap1 GTP/GDP exchanger, resulted in the progressive decline of the numbers of HSPC repopulated in BM with time and impaired long-term hematopoiesis of all cell lineages. C3G-F/HSPC were sustained for months in spleen retaining hematopoietic potential, but these cells inefficiently contributed to overall hematopoietic reconstitution. C3G-F/HSPC showed enhanced proliferation and differentiation with accelerated progenitor cell exhaustion in response to stem cell factor (SCF). Using a Ba/F3 cell line, we confirmed that the increased basal Rap1GTP levels with C3G-F expression caused a markedly prolonged activation of c-Kit receptor and downstream signaling through SCF ligation. A minor population of C3G-F/HSPC also showed enhanced proliferation in the presence of thrombopoietin (TPO) compared to Vect/HSPC. Current results suggest an important role of basal Rap1 activation status of HSPC in their maintenance in BM for sustaining long-term adult hematopoiesis.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal , Proteínas de Unión a Telómeros/metabolismo , Animales , Biomarcadores , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Inmunofenotipificación , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo , Complejo Shelterina , Factor de Células Madre/farmacología , Proteínas de Unión al GTP rap/metabolismo
15.
J Immunol Methods ; 467: 29-36, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30738040

RESUMEN

A clonogenic assay system for thymic epithelial cells (TECs) is of crucial importance for identifying thymic epithelial stem and/or progenitor cells, evaluating their activities, and understanding the mechanisms of thymic involution. However, current systems are not sufficiently sensitive at detecting and quantifying TEC colonies from the adult thymus. Here, we optimized the culture condition to detect visible colonies from adult TECs by modifying our previous culture methods. Epidermal growth factor and leukemia inhibitory factor significantly enhanced the colony-forming efficiency of total TECs from embryo as well as adult mice when added 3 days after plating. Importantly, characteristics of the TEC colonies formed by the improved condition were almost equivalent to those by the original culture condition with respect to self-renewal and the expression of cell surface markers and intracellular keratins. Furthermore, the colonies derived from total TECs showed immature phenotypes and generated both mature cortical TECs and medullary TECs upon implantation in vivo. These data indicate a more sensitive clonogenic assay system for TECs was established and suggest the improved culture condition supports the colony formation of stem/progenitor cells for cTECs, mTECs and/or bipotent TECs.


Asunto(s)
Técnicas de Cultivo de Célula , Células Epiteliales/citología , Timo/citología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL
16.
Int Immunol ; 31(3): 127-139, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30534943

RESUMEN

Hassall's corpuscles (HCs) are composed of cornifying, terminally differentiated medullary thymic epithelial cells (mTECs) that are developed under the control of Aire. Here, we demonstrated that HC-mTECs show features of cellular senescence and produce inflammatory cytokines and chemokines including CXCL5, thereby recruiting and activating neutrophils to produce IL-23 in the thymic medulla. We further indicated that thymic plasmacytoid dendritic cells (pDCs) expressing IL-23 receptors constitutively produced Ifna, which plays a role in single positive (SP) cell maturation, in an Il23a-dependent manner. Neutrophil depletion with anti-Ly6G antibody injection resulted in a significant decrease of Ifna expression in the thymic pDCs, suggesting that thymic neutrophil activation underlies the Ifna expression in thymic pDCs in steady state conditions. A New Zealand White mouse strain showing HC hyperplasia exhibited greater numbers and activation of thymic neutrophils and pDCs than B6 mice, whereas Aire-deficient B6 mice with defective HC development and SP thymocyte maturation showed significantly compromised numbers and activation of these cells. These results collectively suggested that HC-mTECs with cell-senescence features initiate a unique cell activation cascade including neutrophils and pDCs leading to the constitutive IFNα expression required for SP T-cell maturation in the thymic medulla.


Asunto(s)
Senescencia Celular , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interferón-alfa/biosíntesis , Neutrófilos/inmunología , Neutrófilos/metabolismo , Timo/inmunología , Animales , Células Cultivadas , Células Dendríticas/citología , Humanos , Interferón-alfa/metabolismo , Ratones , Ratones Endogámicos , Neutrófilos/citología , Timo/citología
17.
Front Immunol ; 9: 1984, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30279688

RESUMEN

Bile acids (BAs) control metabolism and inflammation by interacting with several receptors. Here, we report that intravenous infusion of taurodeoxycholate (TDCA) decreases serum pro-inflammatory cytokines, normalizes hypotension, protects against renal injury, and prolongs mouse survival during sepsis. TDCA increases the number of granulocytic myeloid-derived suppressor cells (MDSCLT) distinctive from MDSCs obtained without TDCA treatment (MDSCL) in the spleen of septic mice. FACS-sorted MDSCLT cells suppress T-cell proliferation and confer protection against sepsis when adoptively transferred better than MDSCL. Proteogenomic analysis indicated that TDCA controls chromatin silencing, alternative splicing, and translation of the immune proteome of MDSCLT, which increases the expression of anti-inflammatory molecules such as oncostatin, lactoferrin and CD244. TDCA also decreases the expression of pro-inflammatory molecules such as neutrophil elastase. These findings suggest that TDCA globally edits the proteome to increase the number of MDSCLT cells and affect their immune-regulatory functions to resolve systemic inflammation during sepsis.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Sepsis/inmunología , Linfocitos T/inmunología , Ácido Taurodesoxicólico/metabolismo , Animales , Recuento de Células , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Tolerancia Inmunológica , Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oncostatina M/genética , Oncostatina M/metabolismo
18.
Curr Opin Immunol ; 55: 15-21, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30248521

RESUMEN

Many studies describe dysbiosis as a change in the microbiota that accompanies autoimmune illnesses, but little is known about whether these changes are a cause or consequence of an altered immune state. The immune system actively shapes the composition of the microbiota, with divergent outcomes in healthy or autoimmune-prone individuals. The gut microbiota in turn acts as an acquired endocrine organ, influencing the physiology of the host via release of nutrients and chemical messengers. Dysbiosis arising from abnormal immune function can initiate or amplify autoimmunity through multiple mechanisms. We examine how the bidirectional relationship between resident microbes and the immune system contributes to autoimmune diseases.


Asunto(s)
Autoinmunidad/inmunología , Microbioma Gastrointestinal/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Disbiosis/inmunología , Humanos
19.
Eur J Immunol ; 48(10): 1663-1678, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30058200

RESUMEN

Naïve phenotype (NP) T cells spontaneously initiate homeostatic proliferation (HP) as T-cell output is reduced because of physiologic thymic involution with age. However, the effects of sustained HP on overall immune function are poorly understood. We demonstrated that the NP CD8+ T cell population in adult thymectomized mice showing accelerated HP has an increased capacity for TCR-mediated interferon-γ and tumor necrosis factor α production, which is attributed to an increase in CXCR3+ cells in the NP CD8+ T cell population. The CXCR3+ NP CD8+ T cells developed during persistent HP with a slow cell division rate, but rarely during robust antigen-driven proliferation with a fast cell division rate. In ontogeny, the proportions of CXCR3+ cells in the NP CD8+ T cell population showed a biphasic profile, which was high at the newborn and aged stages. Upon transfer, CXCR3+ NP CD8+ T cells, but not CXCR3- NP CD8+ T cells, potently enhanced Th17-mediated inflammatory tissue reactions in vivo. Furthermore, CXCR3high NP CD8+ T cells with similar features were also detected at variable levels in healthy human blood. These results suggest that CXCR3+ NP CD8+ T cells generated during physiological HP significantly impact overall immunity at the immunologically vulnerable neonatal and aged stages.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Interferón gamma/biosíntesis , Receptores CXCR3/genética , Animales , Linfocitos T CD8-positivos/inmunología , División Celular , Células Cultivadas , Citometría de Flujo , Homeostasis , Humanos , Interferón gamma/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células Th17/inmunología
20.
J Leukoc Biol ; 104(2): 253-264, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29603367

RESUMEN

Combined stimulation by IL-2 and IL-18 effectively promotes proliferation of NK cells, whereas singular stimulation does not. In this study, synergistic effects of these cytokines on NK cells proliferation was analyzed, focusing on the roles of IL-18. In splenic resting NK cells from IL-18KO mice, IL-18 rapidly activated NF-κB independently of IL-2, and activated or up-regulated various molecules downstream of PI3K/AKT and mTOR, including S6, Bcl-XL, ATG5, and LC3II, accompanying increases in cell growth and survival. Thus, IL-18 alone was revealed to augment various cellular processes (gene transcription, protein synthesis, survival) in the absence or presence of IL-2. Notably, combined IL-18 and IL-2 promoted autophagosome formation. In addition, priming NK cells with IL-18 augmented IL-2R, especially CD25, and enabled cells to respond to IL-2, resulting in activation of STAT3 and STAT5, followed by increase of cyclin B1 leading to proliferation. However, IL-2 alone failed to activate STAT3 or STAT5 in resting IL18KO NK cells. These results clarify the distinct roles of IL-2 and IL-18 in NK cell proliferation, and the intrinsic roles of IL-18 in various cellular processes, suggesting a range of functions of IL-18 expressed in an array of nonhematopoietic cells.


Asunto(s)
Autofagia/fisiología , Proliferación Celular/fisiología , Interleucina-18/metabolismo , Células Asesinas Naturales/metabolismo , Biosíntesis de Proteínas/fisiología , Animales , Supervivencia Celular/fisiología , Interleucina-2/metabolismo , Activación de Linfocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...