Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nanoscale ; 15(47): 19168-19179, 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-37982186

RESUMO

The status of draining lymph nodes (LNs) is critical for determining the treatment and prognosis of cancer that spreads through the lymphatic system. Indocyanine green (ICG) fluorescence imaging has been widely used in sentinel LN (SLN) biopsy technology and has shown favorable effects. However, this too has its own limitations, such as fluorescence instability and diffusion imaging. In this study, we developed macrophage cell membrane-camouflaged ICG-loaded biomimetic nanoparticles (M@F127-ICG) for accurate SLN imaging. ICG selectively positioned at the hydrophobic-hydrophilic interfaces of pluronic F127 micelles protected itself from quenching in aqueous solution, thereby maintaining fluorescence stability and improving fluorescence intensity. In addition, to further improve the aggregation in SLN, the micellar surface was coated with a layer of biomimetic macrophage cell membrane to target LN-resident macrophages. In vivo fluorescence imaging demonstrated that M@F127-ICG significantly enhanced the fluorescence signal and improved the imaging efficiency of SLN. Thus, selectively positioning ICG in the biomimetic nanoplatform enhanced the fluorescence intensity and stability, providing a novel tracer for timely and accurate SLN imaging.


Assuntos
Linfonodo Sentinela , Linfonodo Sentinela/diagnóstico por imagem , Linfonodo Sentinela/patologia , Verde de Indocianina , Biópsia de Linfonodo Sentinela/métodos , Biomimética , Imagem Óptica/métodos , Micelas , Linfonodos/metabolismo , Corantes/metabolismo
2.
Front Immunol ; 14: 1214675, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37483603

RESUMO

Lysine-specific demethylase 1 (LSD1) is an enzyme that removes lysine methylation marks from nucleosome histone tails and plays an important role in cancer initiation, progression, metastasis, and recurrence. Recent research shows that LSD1 regulates tumor cells and immune cells through multiple upstream and downstream pathways, enabling tumor cells to adapt to the tumor microenvironment (TME). As a potential anti-tumor treatment strategy, immunotherapy has developed rapidly in the past few years. However, most patients have a low response rate to available immune checkpoint inhibitors (ICIs), including anti-PD-(L)1 therapy and CAR-T cell therapy, due to a broad array of immunosuppressive mechanisms. Notably, inhibition of LSD1 turns "cold tumors" into "hot tumors" and subsequently enhances tumor cell sensitivity to ICIs. This review focuses on recent advances in LSD1 and tumor immunity and discusses a potential therapeutic strategy for combining LSD1 inhibition with immunotherapy.


Assuntos
Lisina , Neoplasias , Humanos , Histona Desmetilases/metabolismo , Histonas , Neoplasias/terapia , Imunoterapia , Microambiente Tumoral
3.
J Nanobiotechnology ; 21(1): 13, 2023 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-36639772

RESUMO

Macrophage polarization determines the production of cytokines that fuel the initiation and evolution of rheumatoid arthritis (RA). Thus, modulation of macrophage polarization might represent a potential therapeutic strategy for RA. However, coordinated modulation of macrophages in the synovium and synovial fluid has not been achieved thus far. Herein, we develop a biomimetic ApoA-I mimetic peptide-modified neutrophil membrane-wrapped F127 polymer (R4F-NM@F127) for targeted drug delivery during RA treatment. Due to the high expression of adhesion molecules and chemokine receptors on neutrophils, the neutrophil membrane coating can endow the nanocarrier with synovitis-targeting ability, with subsequent recruitment to the synovial fluid under the chemotactic effects of IL-8. Moreover, R4F peptide modification further endows the nanocarrier with the ability to target the SR-B1 receptor, which is highly expressed on macrophages in the synovium and synovial fluid. Long-term in vivo imaging shows that R4F-NM@F127 preferentially accumulates in inflamed joints and is engulfed by macrophages. After loading of the anti-inflammatory drug celastrol (Cel), R4F-NM@F127-Cel shows a significant reduction in hepatotoxicity, and effectively inhibits synovial inflammation and alleviates joint damage by reprogramming macrophage polarization. Thus, our results highlight the potential of the coordinated targeted modulation of macrophages as a promising therapeutic option for the treatment of RA.


Assuntos
Artrite Reumatoide , Nanopartículas , Humanos , Neutrófilos/metabolismo , Biomimética , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Citocinas , Nanopartículas/uso terapêutico
4.
Front Cell Infect Microbiol ; 12: 1050396, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36506034

RESUMO

Klebsiella pneumoniae (K. pneumoniae) is a typical gram-negative iatrogenic bacterium that often causes bacteremia, pneumonia and urinary tract infection particularly among those with low immunity. Although antibiotics is the cornerstone of anti-infections, the clinical efficacy of ß-lactamase and carbapenems drugs has been weakened due to the emergence of drug-resistant K. pneumoniae. Recent studies have demonstrated that host defense plays a critical role in killing K. pneumoniae. Here, we summarize our current understanding of host immunity mechanisms against K. pneumoniae, including mechanical barrier, innate immune cells, cellular immunity and humoral immunity, providing a theoretical basis and the new strategy for the clinical treatment of K. pneumoniae through improving host immunity.


Assuntos
Antibacterianos , Klebsiella pneumoniae , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bactérias , Carbapenêmicos/farmacologia , Carbapenêmicos/uso terapêutico , beta-Lactamases
5.
Front Pharmacol ; 13: 888588, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35662709

RESUMO

IKKε (inhibitor of nuclear factor kappa-B kinase ε) is a member of the noncanonical NF-κB pathway. It participates in the inflammatory response and innate immunity against bacteria. In recent decades, IKKε has been closely associated with metabolic regulation. Inhibition of the IKKε pathway can improve fat deposition in the liver, reduce subcutaneous fat inflammation, and improve liver gluconeogenesis in obesity. IKKε is expected to be a new therapeutic target for metabolic diseases such as nonalcoholic fatty liver disease, diabetes, and obesity. Herein, we summarize the structural characterization, physiological function, and pathological role of IKKε in metabolic diseases and small molecule inhibitors of IKKε.

6.
Front Cell Infect Microbiol ; 12: 851197, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35651754

RESUMO

Tumor necrosis factor alpha (TNF-α) is a crucial factor in the control of Mycobacterium tuberculosis (Mtb) infection. Pathogenic mycobacteria can inhibit and/or regulate host cell TNF-α production in a variety of ways to evade antituberculosis (anti-TB) immunity as well as facilitate immune escape. However, the mechanisms by which TNF-α expression in host cells is modulated to the benefit of mycobacteria is still an interesting topic and needs further study. Here, we report that macrophages infected with Mycobacterium marinum (Mm)-a close relative of Mtb-upregulated the expression of E3 ubiquitin ligase FBXW7. Specific silencing FBXW7 with small interfering RNA (siRNA) significantly elevates TNF-α expression and eventually promotes the elimination of intracellular bacteria. In turn, overexpression of FBXW7 in Raw264.7 macrophages markedly decreased TNF-α production. Furthermore, partial inhibition of FBXW7 in an Mm-infected murine model significantly reduced TNF-α tissue content, alleviated tissue damage as well as reduced the bacterial load of mouse tails. Finally, FBXW7 could decrease TNF-α in a K63-linked ubiquitin signaling dependent manner. Taken together, our study uncovered a previously unknown role of FBXW7 in regulating TNF-α dynamics during mycobacterial infection, which provides new insights into understanding the role of FBXW7 in anti-tuberculosis immunity and its related clinical significance.


Assuntos
Mycobacterium marinum , Mycobacterium tuberculosis , Animais , Proteína 7 com Repetições F-Box-WD/genética , Proteína 7 com Repetições F-Box-WD/metabolismo , Evasão da Resposta Imune , Camundongos , Mycobacterium marinum/genética , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
7.
Biochim Biophys Acta Mol Basis Dis ; 1867(7): 166130, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33746034

RESUMO

A high level of low-density lipoprotein cholesterol (LDL) is one of the most important risk factors for coronary artery disease (CAD), the leading cause of death worldwide. However, a low concentration of LDL may be protective. Genome-wide association studies revealed that variation in ADTRP gene increased the risk of CAD. In this study, we found that a low concentration of oxidized-LDL induced the expression of ADTRP. Further analyses showed that knockdown of the expression of LDL receptor genes LDLR, CD36, or LOX-1 significantly downregulated ADTRP expression, whereas overexpression of LDLR/CD36/LOX-1 markedly increased ADTRP expression through the NF-κB pathway. Like ADTRP, LDLR, CD36 and LOX-1 were all involved in endothelial cell (EC) functions relevant to the initiation of atherosclerosis. Downregulation of LDLR/CD36/LOX-1 promoted monocyte adhesion to ECs and transendothelial migration of monocytes by increasing expression of ICAM-1, VCAM-1, E-selectin and P-selectin, decreased EC proliferation and migration, and increased EC apoptosis, thereby promoting the initiation of atherosclerosis. Opposite effects were observed with the overexpression of ADTRP and LDLR/CD36/LOX-1 in ECs. Interestingly, through the NF-κB and AKT pathways, overexpression of ADTRP significantly upregulated the expression of LDLR, CD36, and LOX-1, and knockdown of ADTRP expression significantly downregulated the expression of LDLR, CD36, and LOX-1. These data suggest that ADTRP and LDL receptors LDLR/CD36/LOX-1 positively regulate each other, and form a positive regulatory loop that regulates endothelial cell functions, thereby providing a potential protective mechanism against atherosclerosis. Our findings provide a new molecular mechanism by which deregulation of ADTRP and LDLR/CD36/LOX-1 promote the development of atherosclerosis and CAD.


Assuntos
Aterosclerose/patologia , Antígenos CD36/metabolismo , Células Endoteliais/patologia , Retroalimentação Fisiológica , Proteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Receptores Depuradores Classe E/metabolismo , Aterosclerose/metabolismo , Antígenos CD36/genética , Adesão Celular , Doença da Artéria Coronariana , Células Endoteliais/metabolismo , Humanos , Proteínas de Membrana/genética , Monócitos/metabolismo , Monócitos/patologia , Receptores de LDL/genética , Receptores Depuradores Classe E/genética , Migração Transendotelial e Transepitelial
8.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(1): 16-23, 2021 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-33441224

RESUMO

Objective To investigate the changes of subsets of thymocytes, thymic epithelial cells (TECs) and T lymphocytes in the spleen of mice at different growth stages, and to explore the effect of Rho-associated coiled-coil protein kinase (ROCK) inhibitor on thymus regeneration in aged mice. Methods The thymus and spleens were harvested from female C57BL/6 mice at juvenile, mature adult, middle-aged and aged phases. The subsets of thymocytes, TECs and T cells in the spleen were analyzed by flow cytometry (FCM). TECs of aging mice were treated with ROCK inhibitor in vitro. Cell proliferation was observed using fluorescence immune-linked spot analyzer. Aged mice of 20-month old were treated with ROCK inhibitor in vivo. The changes of thymocytes, TECs and T cell subgroups in the spleen were detected with FCM. Results The total numbers of thymocytes and TECs as well as the number of each cell subpopulation decreased significantly with aging. The proportions of CD4+ naive T cells, CD8+ naive T cells and CD4+ recent thymus emigrant cells (RTEs) in the spleen showed significant decreasing trends although there were not obvious changes in the proportions of CD4+ T cells and CD8+ T cells in the spleen of mice during aging. ROCK inhibitor facilitated the proliferation of TECs in aging mice in vitro. ROCK inhibitor also increased the numbers of the subsets of thymocytes, TECs and T cells in the spleen of aged mice significantly. Conclusion The mouse thymus undergoes progressing degeneration with aging. ROCK inhibitor has potential of relieving the atrophy of thymus, facilitating thymus regeneration in aged mice.


Assuntos
Linfócitos T CD8-Positivos , Baço , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Regeneração , Subpopulações de Linfócitos T , Timo , Quinases Associadas a rho
9.
Gene ; 753: 144805, 2020 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-32445923

RESUMO

Genomic variants in both ADTRP and TFPI genes are associated with risk of coronary artery disease (CAD). ADTRP regulates TFPI expression and endothelial cell functions involved in the initiation of atherosclerotic CAD. ADTRP also specifies primitive myelopoiesis and definitive hematopoiesis by upregulating TFPI expression. However, the underlying molecular mechanism is unknown. Here we show that transcription factor POU1F1 is the key by which ADTRP regulates TFPI expression. Luciferase reporter assays, chromatin-immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA) in combination with analysis of large and small deletions of the TFPI promoter/regulatory region were used to identify the molecular mechanism by which ADTRP regulates TFPI expression. Genetic association was assessed using case-control association analysis and phenome-wide association analysis (PhenGWA). ADTRP regulates TFPI expression at the transcription level in a dose-dependent manner. The ADTRP-response element was localized to a 50 bp region between -806 bp and -756 bp upstream of TFPI transcription start site, which contains a binding site for POU1F1. Deletion of POU1F1-binding site or knockdown of POU1F1 expression abolished ADTRP-mediated transcription of TFPI. ChIP and EMSA demonstrated that POU1F1 binds to the ADTRP response element. Genetic analysis identified significant association between POU1F1 variants and risk of CAD. PhenGWA identified other phenotypic traits associated with the ADTRP-POU1F1-TFPI axis such as lymphocyte count (ADTRP), waist circumference (TFPI), and standing height (POU1F1). These data identify POU1F1 as a transcription factor that regulates TFPI transcription in response to ADTRP, and link POU1F1 variants to risk of CAD for the first time.


Assuntos
Doença da Artéria Coronariana/metabolismo , Lipoproteínas/biossíntese , Proteínas de Membrana/metabolismo , Fator de Transcrição Pit-1/metabolismo , Aterosclerose/genética , Estudos de Casos e Controles , Linhagem Celular , Imunoprecipitação da Cromatina/métodos , Doença da Artéria Coronariana/genética , Bases de Dados Genéticas , Células Endoteliais/metabolismo , Genes Homeobox , Células HeLa , Humanos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Regiões Promotoras Genéticas , Elementos de Resposta , Sítio de Iniciação de Transcrição , Transcrição Gênica
10.
Immunol Cell Biol ; 96(2): 229-235, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29363160

RESUMO

Hepatocyte growth factor (HGF) and its receptor c-Met signaling have been implicated in regulating various types of cells including epithelial cells. We have previously reported that c-Met is expressed by thymic epithelial cells (TECs), and that in vivo administration of hybrid cytokines containing IL-7 and the beta- or alpha-chain of HGF significantly increase the number of TECs. In order to study the role of c-Met signaling in TECs, we generated conditional knockout (cKO) mice in which c-Met was specifically deleted in TECs using a Foxn1-Cre transgene. We show here that c-Met deficiency in TECs results in age-progressive reduction in TEC number and reduced number of regulatory T cells. Consequently, c-Met TEC cKO mice displayed an autoimmune phenotype. Thus, c-Met signaling in TECs is important for the maintenance of TECs and immune self-tolerance.


Assuntos
Autoimunidade , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Deleção de Genes , Marcação de Genes , Proteínas Proto-Oncogênicas c-met/genética , Animais , Contagem de Células , Senescência Celular , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-met/deficiência , Proteínas Proto-Oncogênicas c-met/metabolismo , Linfócitos T Reguladores/patologia , Timócitos/patologia , Timo/patologia
11.
Nutrients ; 9(4)2017 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-28425936

RESUMO

Renal ischemia/reperfusion (I/R) injury continues to be a complicated situation in clinical practice. Genistein, the main isoflavone found in soy products, is known to possess a wide spectrum of biochemical and pharmacological activities. However, the protective effect of genistein on renal I/R injury has not been well investigated. In the current study, we explore whether genistein exhibits its renal-protective effects through SIRT1 (Sirtuin 1) in I/R-induced mice model. We found the treatment of genistein significantly reduced renal I/R-induced cell death, simultaneously stimulating renal cell proliferation. Meanwhile, SIRT1 expression was up-regulated following the administration of genistein in renal region. Furthermore, pharmacological inhibition or shRNA-mediated depletion of SIRT1 significantly reversed the protective effect of genistein on renal dysfunction, cellular damage, apoptosis, and proliferation following I/R injury, suggesting an indispensible role of the increased SIRT1 expression and activity in this process. Meanwhile, the reduced p53 and p21 expression and increased PCNA (Proliferating Cell Nuclear Antigen) expression were blocked after the depletion of SIRT1 compared with the genistein treatment group in the renal I/R process. Hence, our results provided further experimental basis for the potential use of genistein for the treatment of kidney disease with deficiency of SIRT1 activity.


Assuntos
Genisteína/farmacologia , Nefropatias/tratamento farmacológico , Rim/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Sirtuína 1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Modelos Animais de Doenças , Rim/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Traumatismo por Reperfusão/complicações , Sirtuína 1/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Stem Cells Transl Med ; 6(1): 121-130, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28170174

RESUMO

Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many malignant and nonmalignant diseases. However, chronic graft-versus-host disease (cGVHD) remains a significant cause of late morbidity and mortality after allogeneic HSCT. cGVHD often manifests as autoimmune syndrome. Thymic epithelial cells (TECs) play a critical role in supporting negative selection and regulatory T-cell (Treg) generation. Studies have shown that damage in TECs is sufficient to induce cGVHD. We have previously reported that mouse embryonic stem cells (mESCs) can be selectively induced to generate thymic epithelial progenitors (TEPs) in vitro. When transplanted in vivo, mESC-TEPs further develop into TECs that support T-cell development. We show here that transplantation of donor-origin mESC-TEPs into cGVHD recipients induces immune tolerance to both donor and host antigens and prevents the development of cGVHD. This is associated with more TECs and Tregs. Our results suggest that embryonic stem cell-derived TEPs may offer a new tool to control cGVHD. Stem Cells Translational Medicine 2017;6:121-130.


Assuntos
Células Epiteliais/transplante , Doença Enxerto-Hospedeiro/prevenção & controle , Células-Tronco Embrionárias Murinas/transplante , Transplante de Células-Tronco , Timo/citologia , Animais , Antígenos/metabolismo , Contagem de Células , Diferenciação Celular , Doença Crônica , Células Epiteliais/citologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/citologia , Linfócitos T Reguladores/citologia , Timócitos/citologia
13.
Br J Haematol ; 175(3): 505-516, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27447780

RESUMO

Given that donor T cells from a transplant contribute both the desired graft-versus-tumour (GVT) effect and detrimental graft-versus-host disease (GVHD), strategies to separate GVHD and GVT activity are a major clinical goal. We have previously demonstrated that in vivo administration of a recombinant (r)IL-7/HGFß hybrid cytokine, consisting of interleukin-7 (IL-7, IL7) and the ß-chain of hepatocyte growth factor (HGFß), significantly inhibits the growth of cancer cells in murine tumour models. The antit-umour effect of rIL-7/HGFß is related to a marked infiltration T cells in the tumour tissues. We have also shown that GVHD was not induced in rIL-7/HGFß-treated T cell-depleted allogeneic haematopoietic stem cell transplantation (HSCT) recipients. We show here that, in T cell-replete allogeneic HSCT murine models, rIL-7/HGFß attenuated acute GVHD (aGVHD), while promoting GVT activity. This was related to an alteration of donor T cell trafficking, with an increased infiltration of donor T cells into tumour tissues and the lympho-haematopoietic system but decreased number of the T cells in the GVHD target organs. Therefore, rIL-7/HGFß may offer a new tool to alleviate aGVHD while prompting GVT, and to study the molecular regulation of T cell trafficking.


Assuntos
Quimiotaxia/efeitos dos fármacos , Doença Enxerto-Hospedeiro/imunologia , Efeito Enxerto vs Tumor/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Interleucina-7/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Doadores de Tecidos , Animais , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Quimiotaxia/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/mortalidade , Efeito Enxerto vs Tumor/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Fator de Crescimento de Hepatócito/genética , Interleucina-7/genética , Camundongos , Neoplasias/complicações , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Proteínas Recombinantes de Fusão/genética , Linfócitos T/metabolismo , Carga Tumoral/efeitos dos fármacos
14.
Mol Cancer Ther ; 15(10): 2413-2421, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27474151

RESUMO

Both IL7 and IL15 have become important candidate immunomodulators for cancer treatment. However, IL7 or IL15 used alone suffers from shortcomings, such as short serum half-life and limited antitumor effect. We have cloned and expressed a recombinant (r) IL7/IL15 fusion protein in which IL7 and IL15 are linked by a flexible linker. We then compared the antitumor effect of rIL7/IL15 with the individual factors rIL7 and/or rIL15. We show here that rIL7/IL15 has a higher antitumor activity than the combination of the individual factors in both murine B16F10 melanoma and CT-26 colon cancer models. This was associated with a significant increase in tumor infiltration of T cells, DCs, and NK cells and a decrease in regulatory T cells (Tregs). In addition, rIL7/IL15-treated DCs had higher expression of costimulatory molecules CD80 and CD86. The higher antitumor activity of rIL7/IL15 is likely due to its longer in vivo half-life and different effects on immune cells. Our results suggest that rIL7/IL15 may offer a new tool to enhance antitumor immunity and treat cancer. Mol Cancer Ther; 15(10); 2413-21. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Interleucina-15 , Interleucina-7 , Proteínas Recombinantes de Fusão/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Interleucina-15/genética , Interleucina-15/farmacologia , Interleucina-7/genética , Interleucina-7/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental , Camundongos , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Eur J Immunol ; 46(6): 1518-28, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27125859

RESUMO

A prolonged period of T-cell recovery is the major challenge in hematopoietic stem cell transplantation (HSCT). Thymic epithelial cells (TECs) are the major component of the thymic microenvironment for T-cell generation. However, TECs undergo degeneration over time. FOXN1 plays a critical role in TEC development and is required to maintain adult TECs for thymopoiesis. To investigate the potential application of FOXN1, we have cloned and expressed recombinant FOXN1 protein (rFOXN1) that was fused with cell-penetrating peptides. We show here that the rFOXN1 protein can translocate from the cell surface into the cytoplasm and nucleus. Administration of rFOXN1 into both congenic and allogeneic HSCT recipient mice increased the number of TECs, resulting in enhanced thymopoiesis that led to an increased number of functional T cells in the periphery. The increased number of TECs is due to the enhanced survival and proliferation of TECs. Our results suggest that rFOXN1 has the potential to be used in enhancing T-cell regeneration in patients following HSCT.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fatores de Transcrição Forkhead/farmacologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Linfopoese/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Linfócitos T/citologia , Animais , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/isolamento & purificação , Camundongos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Regeneração , Timócitos/citologia , Timócitos/efeitos dos fármacos , Timócitos/metabolismo , Timo/citologia , Timo/imunologia
16.
Sci Rep ; 5: 9882, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26044259

RESUMO

Thymic epithelial cells (TECs) are the major components of the thymic microenvironment for T cell development. TECs are derived from thymic epithelial progenitors (TEPs). It has been reported that human ESCs (hESCs) can be directed to differentiate into TEPs in vitro. However, the efficiency for the differentiation is low. Furthermore, transplantation of hESC-TEPs in mice only resulted in a very low level of human T cell development from co-transplanted human hematopoietic precursors. We show here that we have developed a novel protocol to efficiently induce the differentiation of hESCs into TEPs in vitro. When transplanted into mice, hESC-TEPs develop into TECs and form a thymic architecture. Most importantly, the hESC-TECs support the long-term development of functional mouse T cells or a higher level of human T cell development from co-transplanted human hematopoietic precursors. The hESC-TEPs may provide a new approach to prevent or treat patients with T cell immunodeficiency.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Timo/citologia , Animais , Técnicas de Cultura de Células , Células-Tronco Embrionárias Humanas/transplante , Humanos , Técnicas In Vitro , Linfopoese , Camundongos , Camundongos Nus , Transplante de Células-Tronco , Linfócitos T/citologia , Linfócitos T/metabolismo
17.
J Autoimmun ; 58: 36-47, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25618825

RESUMO

Tolerance induction, and thus prevention or treatment of autoimmune disease, is not only associated with the persistent presence of self-antigen in the thymus, but also relies on a functional thymus; however, the thymus undergoes profound age-dependent involution. Thymic epithelial cells (TECs) are the major component of the thymic microenvironment for T cell development. We have reported that mouse embryonic stem cells (mESCs) can be induced in vitro to generate thymic epithelial progenitors (TEPs) that further develop into functional TECs in vivo. We show here that transplantation of mESC-TEPs expressing self-antigen myelin oligodendrocyte glycoprotein (MOG) in mice results in enhanced T cell regeneration, long-term expression of MOG in the thymus, prevention of experimental autoimmune encephalomyelitis (EAE) development, and remission of established EAE. Our findings indicate that transplantation of ESC-TEPs expressing disease-causative self-antigen(s) may provide an effective approach for the prevention and treatment of autoimmune disease.


Assuntos
Células-Tronco Embrionárias/transplante , Encefalomielite Autoimune Experimental/terapia , Esclerose Múltipla/terapia , Glicoproteína Mielina-Oligodendrócito/imunologia , Timo/imunologia , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/imunologia , Células Epiteliais/imunologia , Células Epiteliais/transplante , Feminino , Humanos , Tolerância Imunológica , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/genética , Transplante de Células-Tronco , Timo/citologia
18.
Immunology ; 144(2): 245-53, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25074726

RESUMO

We have reported that in vivo administration of the hybrid cytokine rIL-7/HGFß or rIL-7/HGFα, which contains interleukin-7 (IL-7) and the ß- or α-chain of hepatocyte growth factor (HGF), significantly enhances thymopoiesis in mice after bone marrow transplantation. We have shown that the HGF receptor, c-Met, is involved in the effect of the hybrid cytokines. To address the role of c-Met signalling in thymocyte development and recovery, we generated conditional knockout (cKO) mice in which c-Met was specifically deleted in T cells by crossing c-Met(ft/ft) mice with CD4-Cre transgenic mice. We show here that although the number of total thymocytes and thymocyte subsets in young c-Met cKO mice is comparable to age-matched control (Ctrl) mice, the cKO mice were more susceptible to sub-lethal irradiation and dexamethasone treatment. This was demonstrated by low recovery in thymic cellularity in c-Met cKO mice after insult. Furthermore, the number of total thymocytes and thymocyte subsets was markedly reduced in 6- to 12-month-old cKO mice compared with age-matched Ctrl mice, and the thymic architecture of 12-month-old cKO mice was similar to that of 20-month-old wild-type mice. In addition, c-Met deficiency reduced cell survival and the expression of Bcl-xL in double-positive thymocytes, and decreased cell proliferation and the expression of cyclin E and cyclin-dependent kinase 5 in single-positive thymocytes. Our data indicate that c-Met signalling plays an important role in thymic regeneration after thymic insult. In addition, T-cell-specific inactivation of c-Met accelerates age-related thymic involution.


Assuntos
Diferenciação Celular/imunologia , Proteínas Proto-Oncogênicas c-met/imunologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/citologia , Timo/imunologia , Animais , Anti-Inflamatórios/farmacologia , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Sobrevivência Celular , Ciclina E/biossíntese , Quinase 5 Dependente de Ciclina/biossíntese , Dexametasona/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-met/genética , Regeneração/imunologia , Subpopulações de Linfócitos T/imunologia , Timócitos/efeitos dos fármacos , Timócitos/imunologia , Timócitos/efeitos da radiação , Timo/efeitos dos fármacos , Timo/efeitos da radiação , Proteína bcl-X/biossíntese
19.
Stem Cells Dev ; 23(13): 1491-500, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24564535

RESUMO

The T-box transcriptional factor (Tbx) family of transcriptional factors has distinct roles in a wide range of embryonic differentiation or response pathways. Tbx1, a T-box transcription factor, is an important gene for the human congenital disorder 22q11.2 deletion syndrome. Induced pluripotent stem cell (iPSC) technology offers new opportunities for both elucidation of the pathogenesis of diseases and the development of stem-cell-based therapies. In this study, we generated iPSCs from Tbx1(-/-) and Tbx1(+/+) fibroblasts and investigated the spontaneous differentiation potential of iPSCs by detailed lineage analysis of the iPSC-derived embryoid bodies. Undifferentiated Tbx1(-/-) and Tbx1(+/+) iPSCs showed similar expression levels of pluripotent markers. The ability of the Tbx1(-/-) iPSCs to generate endodermal and mesodermal lineages was compromised upon spontaneous differentiation into embryonic bodies. Restoration of Tbx1 expression in the Tbx1(-/-) iPSCs to normal levels using an inducible lentiviral system rescued these cells from the potential of defective differentiation. Interestingly, overexpression of Tbx1 in the Tbx1(-/-) iPSCs to higher levels than in the Tbx1(+/+) iPSCs again led to a defective differentiation potential. Additionally, we observed that expression of fibroblast growth factor (FGF) 10 and FGF8 was downregulated in the Tbx1(-/-) iPSC-derived cells, which suggests that Tbx1 regulates the expression of FGFs. Taken together, our results implicated the Tbx1 level as an important determinant of endodermal and mesodermal lineage differentiation during embryonic development.


Assuntos
Endoderma/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Mesoderma/citologia , Proteínas com Domínio T/fisiologia , Animais , Diferenciação Celular , Desenvolvimento Embrionário , Fator 10 de Crescimento de Fibroblastos/fisiologia , Fator 8 de Crescimento de Fibroblasto/fisiologia , Expressão Gênica , Técnicas de Inativação de Genes , Camundongos
20.
PLoS One ; 8(12): e82998, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349415

RESUMO

T cell immunodeficiency is a major complication of bone marrow (BM) transplantation (BMT). Therefore, approaches to enhance T cell reconstitution after BMT are required. We have purified a hybrid cytokine, consisting of IL-7 and the ß-chain of hepatocyte growth factor (HGFß) (IL-7/HGFß), from a unique long-term BM culture system. We have cloned and expressed the IL-7/HGFß gene in which the IL-7 and HGFß genes are connected by a flexible linker to generate rIL-7/HGFß protein. Here, we show that rIL-7/HGFß treatment enhances thymopoiesis after allogeneic BMT. Although rIL-7 treatment also enhances the number of thymocytes, rIL-7/HGFß hybrid cytokine was more effective than was rIL-7 and the mechanisms by which rIL-7 and rIL-7/HGFß increase the numbers of thymocytes are different. rIL-7 enhances the survival of double negative (DN), CD4 and CD8 single positive (SP) thymocytes. In contrast, rIL-7/HGFß enhances the proliferation of the DN, SP thymocytes, as well as the survival of CD4 and CD8 double positive (DP) thymocytes. rIL-7/HGFß treatment also increases the numbers of early thymocyte progenitors (ETPs) and thymic epithelial cells (TECs). The enhanced thymic reconstitution in the rIL-7/HGFß-treated allogeneic BMT recipients results in increased number and functional activities of peripheral T cells. Graft-versus-host-disease (GVHD) is not induced in the rIL-7/HGFß-treated BMT mice. Therefore, rIL-7/HGFß may offer a new tool for the prevention and/or treatment of T cell immunodeficiency following BMT.


Assuntos
Transplante de Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Fator de Crescimento de Hepatócito/farmacologia , Interleucina-7/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Aloenxertos , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Fator de Crescimento de Hepatócito/genética , Interleucina-7/genética , Camundongos , Proteínas Recombinantes de Fusão/genética , Recuperação de Função Fisiológica/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...