Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Biomedicines ; 12(1)2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38255242

RESUMO

The function of mucosal-associated invariant T (MAIT) cells, a burgeoning member of innate-like T cells abundant in humans and implicated in many diseases, remains obscure. To explore this, mice with a rearranged T cell receptor (TCR) α or ß locus, specific for MAIT cells, were generated via induced pluripotent stem cells derived from MAIT cells and were designated Vα19 and Vß8 mice, respectively. Both groups of mice expressed large numbers of MAIT cells. The MAIT cells from these mice were activated by cytokines and an agonist to produce IFN-γ and IL-17. While Vß8 mice showed resistance in a cancer metastasis model, Vα19 mice did not. Adoptive transfer of MAIT cells from the latter into the control mice, however, recapitulated the resistance. These mice present an implication for understanding the role of MAIT cells in health and disease and in developing treatments for the plethora of diseases in which MAIT cells are implicated.

2.
Allergol Int ; 73(1): 40-47, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37567833

RESUMO

Despite recent advances in asthma treatments, the search for novel therapies remains necessary because there are still patients with recurrent asthma exacerbations and poor responses to the existing treatments. Since group 2 innate lymphoid cells (ILC2) play a pivotal role in asthma by triggering and exacerbating type 2 inflammation, controlling ILC2s function is key to combating severe asthma. Mucosal-associated invariant T (MAIT) cells are innate-like T cells abundant in humans and are activated both in a T cell receptor-dependent and -independent manner. MAIT cells are composed of MAIT1 and MAIT17 based on the expression of transcription factors T-bet and RORγt, respectively. MAIT cells play pivotal roles in host defense against pathogens and in tissue repair and are essential for the maintenance of immunity and hemostasis. Our recent studies revealed that MAIT cells inhibit both ILC2 proliferation and functions in a mouse model of airway inflammation. MAIT cells may alleviate airway inflammation in two ways, by promoting airway epithelial cell barrier repair and by repressing ILC2s. Therefore, reagents that promote MAIT cell-mediated suppression of ILC2 proliferation and function, or designer MAIT cells (genetically engineered to suppress ILC2s or promote repair of airway damage), may be effective therapeutic agents for severe asthma.


Assuntos
Asma , Células T Invariantes Associadas à Mucosa , Camundongos , Animais , Humanos , Imunidade Inata , Linfócitos , Inflamação
3.
Biomedicines ; 10(12)2022 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-36551916

RESUMO

Mucosal-associated invariant T (MAIT) cells, a burgeoning type of the innate-like T cells, play a crucial role in maintaining immune homeostasis, particularly in host defense. Although many studies have implied the use of MAIT cells in tumor immunity, whether MAIT cells are pro-tumor or anti-tumor has remained elusive, as in the case for other innate-like T cells that possess dichotomous roles in tumor immunity. Although this difficulty persists where endogenous MAIT cells are the target for therapeutic intervention, the advent of induced pluripotent stem-cell-derived MAIT cells (reMAIT cells) will make it possible to harness these cells for immune cell therapy. In this review, we will discuss possible roles of MAIT cells in tumor immunity and the potential of reMAIT cells to treat tumors.

4.
Front Immunol ; 13: 1005226, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36458017

RESUMO

Mucosal-associated invariant T (MAIT) cells, a blossoming member of the innate-like T cells, play a pivotal role in host defense through engaging the mucosal immunity. Although it has been suggested that MAIT cells are somehow implicated in the allergic airway inflammation mediated by group 2 innate lymphoid cells (ILC2s) such as asthma, the precise role(s) of MAIT cells in such inflammation has remained elusive. To explore the possible roles of MAIT cells in the inflammation, we examined whether MAIT cells suppressed the production of T helper (Th) 2 and inflammatory cytokines from ILC2s, and constrained the proliferation of ILC2s, both of which are prerequisite for airway inflammation. Given that laboratory mice are poor at MAIT cells, a novel mouse line rich in MAIT cells was used. We found that mice rich in MAIT cells showed alleviated airway inflammation as evidenced by reduced infiltration of the immune cells and hyperplasia in goblet cells in the lung concomitant with compromised production of Th2 and inflammatory cytokines, while wild type mice exhibited severe inflammation upon challenge with the fungal extracts. In vitro coculture experiments using purified ILC2s and MAIT cells unrevealed that cytokine-stimulated MAIT cells suppressed ILC2s to produce the cytokines as well as to proliferate most likely via production of IFN-γ. Furthermore, reconstitution of the allergic airway inflammation in the highly immunocompromised mice showed that ILC2-mediated inflammation was alleviated in mice that received MAIT cells along with ILC2s. We concluded that MAIT cells played a crucial role in suppressing the cytokine-producing capacity of ILC2s and ILC2 proliferation, that ultimately led to decrease in the allergic airway inflammation. The results open up a novel therapeutic horizon in ILC2-mediated inflammatory diseases by modulating MAIT cell activity.


Assuntos
Células T Invariantes Associadas à Mucosa , Camundongos , Animais , Imunidade Inata , Linfócitos , Inflamação , Citocinas
5.
Elife ; 112022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35379387

RESUMO

Mucosal-associated invariant T (MAIT) cells belong to a family of innate-like T cells that bridge innate and adaptive immunities. Although MAIT cells have been implicated in tumor immunity, it currently remains unclear whether they function as tumor-promoting or inhibitory cells. Therefore, we herein used induced pluripotent stem cell (iPSC) technology to investigate this issue. Murine MAIT cells were reprogrammed into iPSCs and redifferentiated towards MAIT-like cells (m-reMAIT cells). m-reMAIT cells were activated by an agonist in the presence and absence of antigen-presenting cells and MR1-tetramer, a reagent to detect MAIT cells. This activation accompanied protein tyrosine phosphorylation and the production of T helper (Th)1, Th2, and Th17 cytokines and inflammatory chemokines. Upon adoptive transfer, m-reMAIT cells migrated to different organs with maturation in mice. Furthermore, m-reMAIT cells inhibited tumor growth in the lung metastasis model and prolonged mouse survival upon tumor inoculation through the NK cell-mediated reinforcement of cytolytic activity. Collectively, the present results demonstrated the utility and role of m-reMAIT cells in tumor immunity and provide insights into the function of MAIT cells in immunity.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias Pulmonares , Células T Invariantes Associadas à Mucosa , Imunidade Adaptativa , Animais , Células-Tronco Pluripotentes Induzidas/metabolismo , Neoplasias Pulmonares/metabolismo , Camundongos , Mucosa
6.
Methods Mol Biol ; 2098: 237-257, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31792827

RESUMO

Reprogramming differentiated cells into induced pluripotent stem cells (iPSCs) consists in dedifferentiation of the cells into the pluripotent state, i.e., stem cells. Since T cells play a pivotal role in our immune system, T cell reprogramming into iPSCs and subsequent redifferentiation of iPSCs toward the original cells hold a great promise for future cell therapy and for further exploring the biology of such T cells. Mucosal-associated invariant T (MAIT) cells are an innate-like T cells linking innate immunity to adaptive immunity, and believed to be implicated in host protection to infection, in inflammation, and in immune homeostasis, which makes them an attractive target for the clinical intervention. In this chapter, we will outline the protocol for reprogramming MAIT cells to pluripotency with Sendai virus vector and redifferentiation. This technique will allow expansion of MAIT cells for cell therapy against the intractable infectious diseases such as HIV/Tuberculosis or cancer.


Assuntos
Diferenciação Celular/imunologia , Reprogramação Celular/genética , Reprogramação Celular/imunologia , Células T Invariantes Associadas à Mucosa/citologia , Células T Invariantes Associadas à Mucosa/metabolismo , Biomarcadores , Separação Celular/métodos , Transformação Celular Neoplásica , Sangue Fetal/citologia , Imunofluorescência , Expressão Gênica , Imunofenotipagem , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/metabolismo , Reação em Cadeia da Polimerase
7.
Front Immunol ; 8: 1711, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29250077

RESUMO

Although antibiotics to inhibit bacterial growth and small compounds to interfere with the productive life cycle of human immunodeficiency virus (HIV) have successfully been used to control HIV infection, the recent emergence of the drug-resistant bacteria and viruses poses a serious concern for worldwide public health. Despite intensive scrutiny in developing novel antibiotics and drugs to overcome these problems, there is a dilemma such that once novel antibiotics are launched in markets, sooner or later antibiotic-resistant strains emerge. Thus, it is imperative to develop novel methods to avoid this vicious circle. Here, we discuss the possibility of using induced pluripotent stem cell (iPSC)-derived, innate-like T cells to control infection and potential application of these cells for cancer treatment. Mucosal-associated invariant T (MAIT) cells belong to an emerging family of innate-like T cells that link innate immunity to adaptive immunity. MAIT cells exert effector functions without priming and clonal expansion like innate immune cells and relay the immune response to adaptive immune cells through production of relevant cytokines. With these characteristics, MAIT cells are implicated in a wide range of human diseases such as autoimmune, infectious, and metabolic diseases, and cancer. Circulating MAIT cells are often depleted by these diseases and often remain depleted even after appropriate remedy because MAIT cells are susceptible to activation-induced cell death and poor at proliferation in vivo, which threatens the integrity of the immune system. Because MAIT cells have a pivotal role in human immunity, supplementation of MAIT cells into immunocompromised patients suffering from severe depletion of these cells may help recapitulate or recover immunocompetence. The generation of MAIT cells from human iPSCs has made it possible to procure MAIT cells lost from disease. Such technology creates new avenues for cell therapy and regenerative medicine for difficult-to-cure infectious diseases and cancer and contributes to improvement of our welfare.

9.
J Virol ; 91(17)2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28566378

RESUMO

Infant humans and rhesus macaques infected with the human or simian immunodeficiency virus (HIV or SIV), respectively, express higher viral loads and progress more rapidly to AIDS than infected adults. Activated memory CD4+ T cells in intestinal tissues are major primary target cells for SIV/HIV infection, and massive depletion of these cells is considered a major cause of immunodeficiency. Monocytes and macrophages are important cells of innate immunity and also are targets of HIV/SIV infection. We reported previously that a high peripheral blood monocyte turnover rate was predictive for the onset of disease progression to AIDS in SIV-infected adult macaques. The purpose of this study was to determine if earlier or higher infection of monocytes/macrophages contributes to the more rapid progression to AIDS in infants. We observed that uninfected infant rhesus macaques exhibited higher physiologic baseline monocyte turnover than adults. Early after SIV infection, the monocyte turnover further increased, and it remained high during progression to AIDS. A high percentage of terminal deoxynucleotidyltransferase dUTP nick end label (TUNEL)-positive macrophages in the lymph nodes (LNs) and intestine corresponded with an increasing number of macrophages derived from circulating monocytes (bromodeoxyuridine positive [BrdU+] CD163+), suggesting that the increased blood monocyte turnover was required to rapidly replenish destroyed tissue macrophages. Immunofluorescence analysis further demonstrated that macrophages were a significant portion of the virus-producing cells found in LNs, intestinal tissues, and lungs. The higher baseline monocyte turnover in infant macaques and subsequent macrophage damage by SIV infection may help explain the basis of more rapid disease progression to AIDS in infants.IMPORTANCE HIV infection progresses much more rapidly in pediatric cases than in adults; however, the mechanism for this difference is unclear. Using the rhesus macaque model, this work was performed to address why infants infected with SIV progress more quickly to AIDS than do adults. Earlier we reported that in adult rhesus macaques, increasing monocyte turnover reflected tissue macrophage damage by SIV and was predictive of terminal disease progression to AIDS. Here we report that uninfected infant rhesus macaques exhibited a higher physiological baseline monocyte turnover rate than adults. Furthermore, once infected with SIV, infants displayed further increased monocyte turnover that may have facilitated the accelerated progression to AIDS. These results support a role for monocytes and macrophages in the pathogenesis of SIV/HIV and begin to explain why infants are more prone to rapid disease progression.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Infecções por HIV/imunologia , Infecções por HIV/patologia , Humanos , Macaca mulatta/virologia , Macrófagos/virologia , Monócitos/virologia , RNA Viral/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Carga Viral
10.
PLoS One ; 12(3): e0174699, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28346544

RESUMO

Mucosal-associated invariant T cells (MAITs) are innate-like T cells that play a pivotal role in the host defense against infectious diseases, and are also implicated in autoimmune diseases, metabolic diseases, and cancer. Recent studies have shown that induced pluripotent stem cells (iPSCs) derived from MAITs selectively redifferentiate into MAITs without altering their antigen specificity. Such a selective differentiation is a prerequisite for the use of MAITs in cell therapy and/or regenerative medicine. However, the molecular mechanisms underlying this phenomenon remain unclear. Here, we performed methylome and transcriptome analyses of MAITs during the course of differentiation from iPSCs. Our multi-omics analyses revealed that recombination-activating genes (RAG1 and RAG2) and DNA nucleotidylexotransferase (DNTT) were highly methylated with their expression being repressed throughout differentiation. Since these genes are essential for V(D)J recombination of the T cell receptor (TCR) locus, this indicates that nascent MAITs are kept from further rearrangement that may alter their antigen specificity. Importantly, we found that the repression of RAGs was assured in two layers: one by the modulation of transcription factors for RAGs, and the other by DNA methylation at the RAG loci. Together, our study provides a possible explanation for the unaltered antigen specificity in the selective differentiation of MAITs from iPSCs.


Assuntos
Epigênese Genética , Inativação Gênica , Células-Tronco Pluripotentes Induzidas/citologia , Células T Invariantes Associadas à Mucosa/citologia , Recombinação V(D)J/genética , Diferenciação Celular , Metilação de DNA , Perfilação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Células T Invariantes Associadas à Mucosa/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transcriptoma
11.
Springerplus ; 5(1): 1259, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27536542

RESUMO

BACKGROUND: Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination, gliosis and axonal loss in the Central Nervous System. Although the etiology of the disease has remained enigmatic, recent studies have suggested a role of the innate-like T cells, called Mucosal Associated Invariant T cells (MAITs) in the pathophysiology. In the present study, we have analyzed the relative frequency of MAITs and the expression of the cell surface antigens in MAITs to seek a possible link to the disease. RESULTS: There was little difference in the frequency of total MAITs between healthy donors (HDs) and untreated MS patients, whereas the latter harbored more CD8(lo/neg) (DN) MAITs concomitant with a decrease in CD8(high) MAITs and in CD4 MAITs compared with those in HDs. While the expression of CCR5, CCR6, CD95, CD127, and CD150 has increased in untreated subjects compared with that in HDs, CD45RO has declined in untreated subjects in both DN MAITs and CD8(hi) MAITs. FTY720 therapy has increased the relative frequency of total MAITs in a time-dependent fashion up to 2 years. Intriguingly, FTY720 therapy for 3 years reversed the above phenotype, engendering more CD8(high) MAITs accompanied with decreased DN MAITs. FTY720 therapy affected the cytokine production from CD4 T cells and also enhanced the relative frequency of cells producing both TNF-α and IFN-γ from MAITs, CD8 T cells, and CD4 T cells compared with that in untreated subjects. CONCLUSIONS: FTY 720 therapy enhanced the relative frequency of MAITs in MS patients in a time-dependent manner. Although the expression of CD8 in MAITs has been affected early by FTY720, longer treatment has reversed the phenotypic change. These data demonstrated that FTY720 induced dynamic change in the relative frequency and in the phenotype of MAITs in MS.

12.
World J Stem Cells ; 8(4): 158-69, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27114747

RESUMO

Mice have frequently been used to model human diseases involving immune dysregulation such as autoimmune and inflammatory diseases. These models help elucidate the mechanisms underlying the disease and in the development of novel therapies. However, if mice are deficient in certain cells and/or effectors associated with human diseases, how can their functions be investigated in this species? Mucosal-associated invariant T (MAIT) cells, a novel innate-like T cell family member, are a good example. MAIT cells are abundant in humans but scarce in laboratory mice. MAIT cells harbor an invariant T cell receptor and recognize nonpeptidic antigens vitamin B2 metabolites from bacteria and yeasts. Recent studies have shown that MAIT cells play a pivotal role in human diseases such as bacterial infections and autoimmune and inflammatory diseases. MAIT cells possess granulysin, a human-specific effector molecule, but granulysin and its homologue are absent in mice. Furthermore, MAIT cells show poor proliferation in vitro. To overcome these problems and further our knowledge of MAIT cells, we have established a method to expand MAIT cells via induced pluripotent stem cells (iPSCs). In this review, we describe recent advances in the field of MAIT cell research and our approach for human disease modeling with iPSC-derived MAIT cells.

13.
Nihon Rinsho ; 74(12): 2064-2069, 2016 12.
Artigo em Japonês | MEDLINE | ID: mdl-30550668

RESUMO

Nowadays, antibiotic resistance is a serious global health concern, for it is observed every- where on the earth. While antibiotic is effective for controlling pathogens, an inappropriate use of the antibiotic leads to antibiotic resistance. Given that the ability to develop novel anti- biotics is quite limited, a new strategy must be developed to fight against it. Mucosal- associated invariant T cells (MAITs) belong to a family of the innate-like T cells that bridges the gap between the innate and the adaptive immunity. We have generated iPSCs from MAITs and redifferentiated MAITs from the iPSCs. These MAITs exerted anti- mycobacterial activity in mice. Advent of such cells will pave the way to exploit a novel arsen- al against antibiotic resistance.


Assuntos
Células-Tronco Pluripotentes Induzidas , Controle de Infecções , Infecções/terapia , Animais , Humanos , Camundongos
15.
PLoS One ; 10(4): e0121124, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25853812

RESUMO

BACKGROUND: Fibromyalgia (FM) is defined as a widely distributed pain. While many rheumatologists and pain physicians have considered it to be a pain disorder, psychiatry, psychology, and general medicine have deemed it to be a syndrome (FMS) or psychosomatic disorder. The lack of concrete structural and/or pathological evidence has made patients suffer prejudice that FMS is a medically unexplained symptom, implying inauthenticity. Furthermore, FMS often exhibits comorbidity with rheumatoid arthritis (RA) or spondyloarthritis (SpA), both of which show similar indications. In this study, disease specific biomarkers were sought in blood samples from patients to facilitate objective diagnoses of FMS, and distinguish it from RA and SpA. METHODS: Peripheral blood mononuclear cells (PBMCs) from patients and healthy donors (HD) were subjected to multicolor flow cytometric analysis. The percentage of mucosal-associated invariant T (MAIT) cells in PBMCs and the mean fluorescent intensity (MFI) of cell surface antigen expression in MAIT cells were analyzed. RESULTS: There was a decrease in the MAIT cell population in FMS, RA, and SpA compared with HD. Among the cell surface antigens in MAIT cells, three chemokine receptors, CCR4, CCR7, and CXCR1, a natural killer (NK) receptor, NKp80, a signaling lymphocyte associated molecule (SLAM) family, CD150, a degrunulation marker, CD107a, and a coreceptor, CD8ß emerged as potential biomarkers for FMS to distinguish from HD. Additionally, a memory marker, CD44 and an inflammatory chemokine receptor, CXCR1 appeared possible markers for RA, while a homeostatic chemokine receptor, CXCR4 deserved for SpA to differentiate from FMS. Furthermore, the drug treatment interruption resulted in alternation of the expression of CCR4, CCR5, CXCR4, CD27, CD28, inducible costimulatory molecule (ICOS), CD127 (IL-7 receptor α), CD94, NKp80, an activation marker, CD69, an integrin family member, CD49d, and a dipeptidase, CD26, in FMS. CONCLUSIONS: Combined with the currently available diagnostic procedures and criteria, analysis of MAIT cells offers a more objective standard for the diagnosis of FMS, RA, and SpA, which exhibit multifaceted and confusingly similar clinical manifestations.


Assuntos
Artrite Reumatoide/diagnóstico , Fibromialgia/diagnóstico , Fibromialgia/imunologia , Espondilartrite/diagnóstico , Linfócitos T/metabolismo , Antígenos de Superfície/metabolismo , Biomarcadores/sangue , Contagem de Células , Diagnóstico Diferencial , Feminino , Fibromialgia/sangue , Fibromialgia/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Linfócitos T/patologia
17.
Cell Stem Cell ; 12(5): 546-58, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23523177

RESUMO

Mucosal-associated invariant T (MAIT) cells play an important physiological role in host pathogen defense and may also be involved in inflammatory disorders and multiple sclerosis. The rarity and inefficient expansion of these cells have hampered detailed analysis and application. Here, we report an induced pluripotent stem cell (iPSC)-based reprogramming approach for the expansion of functional MAIT cells. We found that human MAIT cells can be reprogrammed into iPSCs using a Sendai virus harboring standard reprogramming factors. Under T cell-permissive conditions, these iPSCs efficiently redifferentiate into MAIT-like lymphocytes expressing the T cell receptor Vα7.2, CD161, and interleukin-18 receptor chain α. Upon incubation with bacteria-fed monocytes, the derived MAIT cells show enhanced production of a broad range of cytokines. Following adoptive transfer into immunocompromised mice, these cells migrate to the bone marrow, liver, spleen, and intestine and protect against Mycobacterium abscessus. Our findings pave the way for further functional analysis of MAIT cells and determination of their therapeutic potential.


Assuntos
Diferenciação Celular , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Mucosa/citologia , Linfócitos T/citologia , Animais , Diferenciação Celular/genética , Proliferação de Células , Feminino , Sangue Fetal/citologia , Regulação da Expressão Gênica , Humanos , Hospedeiro Imunocomprometido/imunologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos SCID , Mucosa/metabolismo , Mycobacterium/imunologia , Infecções por Mycobacterium/imunologia , Infecções por Mycobacterium/prevenção & controle , Linfócitos T/metabolismo
18.
Horm Mol Biol Clin Investig ; 10(2): 249-53, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25436681

RESUMO

Abstract Somatic cell nuclear transfer allows the generation of cloned embryonic stem cells (ESC) and cloned mice from natural killer T (NKT) cells, an innate-type invariant T cell. The progeny of these cloned mice harboring a rearranged T cell receptor α loci specific for NKT cells, Vα14-Jα18, possess an increased number of NKT cells in the primary as well as in the secondary lymphoid organs. NKT cells in these mice are able to produce interferon-γ (IFN-γ) and interleukin-4 (IL-4) when stimulated with dendritic cells (DC) pulsed with an NKT cell agonist, α-galactosylceramide (α-GalCer). The directed differentiation of cloned ESC toward T cells results in quasi-exclusive generation of NKT cells. These NKT cells are functional as evidenced by the production of cytokines such as IFN-γ, IL-4, IL-10, and IL-13 in response to α-GalCer. Furthermore, they mature autonomously in vivo upon adoptive transfer, and exhibit an antigen-specific adjuvant effect, resulting in IFN-γ production from CD8+ T cells. This effect is evident when the growth of tumors is inhibited in an antigen-dependent manner upon tumor inoculation into the mouse. Unfortunately, in humans, NKT cells are rare, and there is no guarantee that the same technique will be applicable for use in the clinic. We, therefore, exploited a different type of T cells in humans. We established induced pluripotent stem cells (iPSC) from these T cells, and succeeded in directed differentiation of the iPSC into monoclonal T cells. The availability of iPSC-derived monoclonal T cells paves the way for their use in regenerative medicine. In addition, they will be useful for drug screening to target unmet medical needs such as autoimmunity, severe infection, allergy, and cancer.

19.
Environ Health Prev Med ; 16(4): 247-52, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21431790

RESUMO

OBJECTIVE: The metabolic syndrome is an important social problem affecting many people in developed countries. Obesity is a leading cause of this syndrome, hence understanding molecular mechanisms underlying obesity is of prime importance for preventive medicine to develop novel methods to alleviate the corresponding social cost as well as for pharmaceutical companies to develop antimetabolic drugs. METHODS: Since adipocytes play an important role in obesity, we explored the signaling pathways leading to differentiation of adipocytes. We used a preadipocyte cell line to monitor the differentiation of adipocytes, and virus-mediated gene transfer to assess the role of the transcription factor Stat5 in adipogenesis. Adipocyte differentiation was assessed by Northern blot and Western blot analyses as well as accumulation of fat droplets in cells. Promoter activity of the proadipogenic transcription factor peroxisome proliferator-activated receptor-gamma (PPARγ) was evaluated by luciferase assay. RESULTS: Virus-mediated gene transfer of the constitutively active form of both Stat5A and Stat5B resulted in enhanced adipocyte differentiation in the absence of fetal bovine serum (FBS) as judged by expression of proadipogenic factors as well as accumulation of fat droplets in cells. Such a proadipogenic effect of Stat5 is, in part, mediated by its ability to enhance transcription of PPARγ, a master transcriptional regulator in adipogenesis. CONCLUSION: The constitutively active form of Stat5A and Stat5B promoted adipocyte differentiation in the absence of FBS via induction of PPARγ.


Assuntos
Adipócitos/fisiologia , Adipogenia , PPAR gama/metabolismo , Fator de Transcrição STAT5/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Animais , Sangue Fetal , Humanos , Camundongos , Transfecção
20.
J Biol Chem ; 285(48): 37884-94, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20876580

RESUMO

Leptin, the product of the ob gene, plays important roles in the regulation of food intake and body weight through its receptor in the hypothalamus. To identify novel transcripts induced by leptin, we performed cDNA subtraction based on selective suppression of the polymerase chain reaction by using mRNA prepared from the forebrain of leptin-injected ob/ob mice. One of the genes isolated was a mouse homolog of human negative regulatory element-binding protein (NREBP). Its expression was markedly increased by leptin in the growth hormone secretagogue-receptor (GHS-R)-positive neurons of the arcuate nucleus and ventromedial hypothalamic nucleus. The promoter region of GHS-R contains one NREBP binding sequence, suggesting that NREBP regulates GHS-R transcription. Luciferase reporter assays showed that NREBP repressed GHS-R promoter activity in a hypothalamic neuronal cell line, GT1-7, and its repressive activity was abolished by the replacement of negative regulatory element in GHS-R promoter. Overexpression of NREBP reduced the protein expression of endogenous GHS-R without affecting the expression of ob-Rb in GT1-7 cells. To determine the functional importance of NREBP in the hypothalamus, we assessed the effects of NREBP on ghrelin action. Although phosphorylation of AMP-activated protein kinase α (AMPKα) was induced by ghrelin in GT1-7 cells, NREBP repressed ghrelin-induced AMPKα phosphorylation. These results suggest that leptin-induced NREBP is an important regulator of GHS-R expression in the hypothalamus and provides a novel molecular link between leptin and ghrelin signaling.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Grelina/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/genética , Transdução de Sinais , Sequência de Aminoácidos , Animais , Linhagem Celular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Grelina/genética , Hipotálamo/citologia , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Grelina/genética , Receptores de Grelina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...