Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Immunol ; 200(6): 1982-1987, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29440507

RESUMO

B6.SJL-Ptprca Pepcb /Boy (CD45.1) mice have been used in hundreds of congenic competitive transplants, with the presumption that they differ from C57BL/6 mice only at the CD45 locus. In this study, we describe a point mutation in the natural cytotoxicity receptor 1 (Ncr1) locus fortuitously identified in the CD45.1 strain. This point mutation was mapped at the 40th nucleotide of the Ncr1 locus causing a single amino acid mutation from cysteine to arginine at position 14 from the start codon, resulting in loss of NCR1 expression. We found that these mice were more resistant to CMV due to a hyper innate IFN-γ response in the absence of NCR1. In contrast, loss of NCR1 increased susceptibility to influenza virus, a result that is consistent with the role of NCR1 in the recognition of influenza Ag, hemagglutinin. This work sheds light on potential confounding experimental interpretation when this congenic strain is used as a tool for tracking lymphocyte development.


Assuntos
Antígenos Ly/genética , Antígenos Comuns de Leucócito/genética , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Mutação Puntual/genética , Animais , Hemaglutininas/imunologia , Imunidade Inata , Interferon gama/imunologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/genética
2.
Gut ; 67(5): 847-859, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28341747

RESUMO

BACKGROUND: Interactions between host immune cells and gut microbiota are crucial for the integrity and function of the intestine. How these interactions regulate immune cell responses in the intestine remains a major gap in the field. AIM: We have identified the signalling lymphocyte activation molecule family member 4 (SLAMF4) as an immunomodulator of the intestinal immunity. The aim is to determine how SLAMF4 is acquired in the gut and what its contribution to intestinal immunity is. METHODS: Expression of SLAMF4 was assessed in mice and humans. The mechanism of induction was studied using GFPtg bone marrow chimaera mice, lymphotoxin α and TNLG8A-deficient mice, as well as gnotobiotic mice. Role in immune protection was revealed using oral infection with Listeria monocytogenes and Cytobacter rodentium. RESULTS: SLAMF4 is a selective marker of intestinal immune cells of mice and humans. SLAMF4 induction occurs directly in the intestinal mucosa without the involvement of the gut-associated lymphoid tissue. Gut bacterial products, particularly those of gut anaerobes, and gut-resident antigen-presenting cell (APC) TNLG8A are key contributors of SLAMF4 induction in the intestine. Importantly, lack of SLAMF4 expression leads the increased susceptibility of mice to infection by oral pathogens culminating in their premature death. CONCLUSIONS: SLAMF4 is a marker of intestinal immune cells which contributes to the protection against enteric pathogens and whose expression is dependent on the presence of the gut microbiota. This discovery provides a possible mechanism for answering the long-standing question of how the intertwining of the host and gut microbial biology regulates immune cell responses in the gut.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas/fisiologia , Mucosa Intestinal/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Animais , Citometria de Fluxo , Vida Livre de Germes , Humanos , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Simbiose
3.
Proc Natl Acad Sci U S A ; 112(42): 13081-6, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26438862

RESUMO

Epstein-Barr virus (EBV) infection causes both Hodgkin's lymphoma (HL) and non-Hodgkin's lymphoma (NHL). The present study reveals that EBV-induced HL and NHL are intriguingly associated with a repopulated immune cell profile in humanized mice. Newborn immunodeficient NSG mice were engrafted with human cord blood CD34(+) hematopoietic stem cells (HSCs) for a 8- or 15-wk reconstitution period (denoted (8w)hN and (15w)hN, respectively), resulting in human B-cell and T-cell predominance in peripheral blood cells, respectively. Further, novel humanized mice were established via engraftment of hCD34(+) HSCs together with nonautologous fetal liver-derived mesenchymal stem cells (MSCs) or MSCs expressing an active notch ligand DLK1, resulting in mice skewed with human B or T cells, respectively. After EBV infection, whereas NHL developed more frequently in B-cell-predominant humanized mice, HL was seen in T-cell-predominant mice (P = 0.0013). Whereas human splenocytes from NHL-bearing mice were positive for EBV-associated NHL markers (hBCL2(+), hCD20(+), hKi67(+), hCD20(+)/EBNA1(+), and EBER(+)) but negative for HL markers (LMP1(-), EBNA2(-), and hCD30(-)), most HL-like tumors were characterized by the presence of malignant Hodgkin's Reed-Sternberg (HRS)-like cells, lacunar RS (hCD30(+), hCD15(+), IgJ(-), EBER(+)/hCD30(+), EBNA1(+)/hCD30(+), LMP(+)/EBNA2(-), hCD68(+), hBCL2(-), hCD20(-/weak,) Phospho STAT6(+)), and mummified RS cells. This study reveals that immune cell composition plays an important role in the development of EBV-induced B-cell lymphoma.


Assuntos
Herpesvirus Humano 4/patogenicidade , Linfócitos/classificação , Linfoma de Células B/imunologia , Linfoma de Células B/virologia , Animais , Humanos , Linfoma de Células B/classificação , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
4.
Proc Natl Acad Sci U S A ; 111(35): 12841-6, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25143585

RESUMO

Invariant natural killer T (iNKT) cells to date represent the best example of cells known to have a hybrid function, representing both innate and adaptive immunity. Shared phenotypic similarities with NK cells together with a rapid response to a cytokine stimulus and a productive TCR engagement are the features that underline the hybrid nature of iNKT cells. Using these criteria, we provide molecular and functional evidence demonstrating that CD1d-independent (CD1d(ind)) NKT cells, a population of CD1d-unrestricted NKT cells, are endowed with a hybrid function far superior to that of iNKT cells: (i) an extensive shared program with NK cells, (ii) a closer Euclidian distance with NK cells, and (iii) the ability to respond to innate stimuli (Poly:IC) with cytotoxic potential in the same manner as NK cells identify a hybrid feature in CD1d(ind)NKT cells that truly fulfills the dual function of an NK and a T cell. Our finding that CD1d(ind)NKT cells are programmed to act like NK cells in response to innate signals while being capable of adaptive responses is unprecedented, and thus might reemphasize CD1d-unrestricted NKT cells as a subset of lymphocytes that could affect biological processes of antimicrobial and tumor immunity in a unique way.


Assuntos
Imunidade Adaptativa/imunologia , Antígenos CD1d/imunologia , Células T Matadoras Naturais/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos CD1d/genética , Linhagem da Célula/imunologia , Feminino , Genômica , Granzimas/imunologia , Imunofenotipagem , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Clin Immunol ; 161(2): 308-15, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26360254

RESUMO

We previously generated humanized TB34N mice that received human fetal thymus (T), bone tissue (B) and fetal liver-derived (FL)-CD34(+) cells (34) in immunodeficient, NOD/SCID IL2Rγ(null) (N) mice. Although humanized TB34N mice had excellent hematopoiesis, here, we sought to further improve this model by additional transplantation of human spleen tissue (S) as a secondary hematopoietic tissue (TBS34N). The human spleen grafts were enlarged and differentiated into a similar morphology of adult humans, including follicular lymphoid structures with T- and B-cells. The TBS34N mice mimicked mature human immune system (HIS): mature T- and B-cells and follicular dendritic cells; activated germinal center B-cells expressing CD71, BR3(+) cells, memory B-cells and activation-induced cytidine deaminase(+) B-cells; CD138(+) plasma cells were enriched in the mouse spleen. HBsAg-specific hIgG antibodies were secreted into the sera of all TBS34N mice upon immunization with HBsAg. Taken together, the humanized TBS34N mice improved mature HIS and achieved adaptive antibody responses.


Assuntos
Linfócitos B/imunologia , Transplante de Tecido Fetal/métodos , Baço/imunologia , Baço/transplante , Imunidade Adaptativa/imunologia , Animais , Formação de Anticorpos/imunologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Linfócitos B/metabolismo , Células Dendríticas Foliculares/imunologia , Citometria de Fluxo , Hematopoese/imunologia , Antígenos de Superfície da Hepatite B/imunologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Modelos Animais , Receptores da Transferrina/imunologia , Receptores da Transferrina/metabolismo , Linfócitos T/imunologia , Transplante Heterólogo
6.
Clin Immunol ; 157(2): 156-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25725428

RESUMO

Both the thymus (T) and bone (B) are necessary hematopoietic niches in adult humans. We previously showed that co-transplantation of human fetal T and B tissues into neonatal immunodeficient NOD/SCID IL2Rγ(null) (NSG, N) mice facilitated hematopoiesis. However, transplantation into neonatal mice resulted in high frequency of early death, making it unrealistic for repetitive experiments. In this study, young adult N mice were pre-engrafted with T and B, T alone, B alone or no tissues. The animals were irradiated and injected with autologous fetal liver (FL)-derived CD34(+) cells (34). The resultant mice were TB34N, T34N, B34N and 34N, respectively, and challenged with T cell dependent antigens (Ags). The humanized TB34N mice showed best performance of these mouse models in many aspects resembling the adult human Ag-experienced spleen. The TB34N mice exhibited better hematopoietic reconstitution; balanced development of T- and B-cell, and common progenitor cells; follicular lymphoid structures with a functional germinal center (GC) enriched with follicular dendritic cells (FDCs) and plasma cells (PCs); secretion of hIgG in the sera in response to Ags at comparable levels to those of human; derivations of hIgG mAb-secreting hybridoma clones. Collectively, the humanized TB34N mice could develop an adaptive immunity that was capable of producing Ag-specific hIgG at a significant level via class switching. This unprecedented TB34N platform in humanized mice would be useful in dissecting human immunity, for generating human Abs and clinical applications.


Assuntos
Imunidade Adaptativa/imunologia , Anticorpos/imunologia , Antígenos CD34/metabolismo , Transplante Ósseo , Transplante de Tecido Fetal , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Baço/imunologia , Timo/transplante , Animais , Formação de Anticorpos , Hematopoese , Xenoenxertos , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Baço/patologia
7.
Biomark Res ; 12(1): 52, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38816856

RESUMO

Protein tyrosine kinase 2 (PTK2), epidermal growth factor receptor (EGFR), and toll-like receptor (TLRs) are amplified in non-small cell lung cancer (NSCLC). However, the functional and clinical associations between them have not been elucidated yet in NSCLC. By using microarray data of non-small cell lung cancer (NSCLC) tumor tissues and matched normal tissues of 42 NSCLC patients, the genetic and clinical associations between PTK2, EGFR, and TLRs were analyzed in NSCLC patients. To verify the functional association, we generated PTK2-knockout (PTK2-KO) lung cancer cells by using CRISPR-Cas9 gene editing method, and performed in vitro cancer progression assay, including 3D tumor spheroid assay, and in vivo xenografted NSG (NOD/SCID/IL-2Rγnull) mouse assay. Finally, therapeutic effects targeted to PTK2 in lung cancer in response to EGF and TLR agonists were verified by using its inhibitor (Defactinib). In summary, we identified that up-regulated PTK2 might be a reliable marker for EGFR- or TLRs-induced lung cancer progression in NSCLC patients via the regulation of the cross-talk between EGFR- and TLRs-mediated signaling. This study provides a theoretical basis for the therapeutic intervention of PTK2 targeting EGFR- or TLRs-induced lung cancer progression.

8.
Am J Pathol ; 180(1): 351-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22062221

RESUMO

Ex vivo expansion of CD34(+) stem cells in contact culture between hCD34(+)CD38(-)Lin(-) cord blood stem cells and human delta-like-expressing AFT024 feeder cells revealed increased amounts of stemness-related proteins such as HoxB4, GATA2, Bmi-1, and p21 and anti-apoptotic proteins such as Bcl-2, Bcl-xL, Mcl-1, and phospho-Bad, when compared with control or noncontact culture. Production of human IL-6 (hIL-6) was markedly elevated in the culture, but was profoundly inhibited by treatment with γ-secretase inhibitor. In addition, Notch-induced activation of STAT3 was directly involved in gene expression of hIL-6 and soluble hIL-6Rα, indicating the close linkage between Notch signaling and hIL-6 production. Furthermore, depletion of soluble hIL-6 (with hIL-6-specific antibodies) and inhibition of IL-6-mediated signals (with a Jak1 inhibitor and wortmannin) severely affected the maintenance of self-renewal of hCD34(+) cord blood cells. It was also observed that the ex vivo expanded CD34(+) cord blood cells were induced to reconstitute human immune cells in nonobese diabetic mice with severe combined immunodeficiency when compared with freshly isolated CD34(+) cord blood cells. Together, these results strongly demonstrate that Notch signaling in the "cell-to-cell contact" between hCD34(+) cord blood and delta-like-expressing AFT024 feeder cells facilitates maintenance of self-renewal of hCD34(+) cord blood cells through direct regulation of hIL-6 production.


Assuntos
Antígenos CD34/metabolismo , Células Sanguíneas/imunologia , Sangue Fetal/citologia , Interleucina-6/biossíntese , Receptores Notch/fisiologia , Animais , Proliferação de Células , Humanos , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 1/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia
9.
Stem Cell Res Ther ; 14(1): 283, 2023 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-37794417

RESUMO

BACKGROUNDS: Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease characterized by synovial inflammation-mediated progressive destruction of the cartilage and bone, resulting in reduced quality of life. We primed human telomerase reverse transcriptase-overexpressing immortalized human adipose tissue-derived mesenchymal stem cells (iMSCs) with serum derived from a non-human primate RA model and studied the immunomodulatory ability of exosomes obtained from primed iMSCs. METHODS: After immunophenotyping, nanoparticle tracking analysis, and in vitro functional tests, Dulbecco's phosphate-buffered saline (dPBS, Group C), exosomes derived from the supernatant of iMSCs (Exo-FBS, Group E), exosomes derived from the supernatant of iMSCs primed with RA serum (Exo-RA, Group F), and methotrexate (Group M) were administered in collagen-induced arthritis (CIA) model mice. dPBS was administered to the normal (N) group for comparison (n = 10/group). RESULTS: Exo-RA had a significantly higher number of exosomes compared to Exo-FBS when measured with nanoparticle tracking analysis or exosome marker CD81, and Transforming growth factor-ß1 amounts were significantly higher in Exo-RA than in Exo-FBS. When Exo-FBS or Exo-RA was administered to the collagen-induced arthritis model, serum interleukin (IL)-4 and the proportion of Th2 (CD4+CD25+GATA3+) and M2 (CD11c - CD206+ of CD45+CD64+) cells were significantly increased compared to the control group. Furthermore, Exo-RA could alleviate cartilage damage by significantly lowering the concentrations of proinflammatory cytokines such as tumor necrosis factor-α, keratinocyte chemoattractant, and IL-12p70. CONCLUSION: Exosomes derived from disease-condition-serum-primed iMSCs ameliorated cartilage damage in a RA model by enhancing TGF-ß1 production, inducing Th2 and M2 polarization and lowering proinflammatory cytokines, TNF-α, KC, and IL-12p70 in the host. Patient-derived serum can be used as an iMSC priming strategy in iMSC-derived exosome treatment of RA.


Assuntos
Artrite Experimental , Artrite Reumatoide , Exossomos , Células-Tronco Mesenquimais , Humanos , Animais , Camundongos , Artrite Experimental/terapia , Fator de Crescimento Transformador beta1/genética , Exossomos/patologia , Qualidade de Vida , Modelos Animais de Doenças , Artrite Reumatoide/terapia , Citocinas , Fator de Necrose Tumoral alfa , Células-Tronco Mesenquimais/patologia
10.
Transl Oncol ; 15(1): 101250, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34688043

RESUMO

Herein, we aimed to elucidate the molecular and cellular mechanism in which ubiquitin-specific protease 8 (USP8) is implicated in liver cancer progression via TRAF6-mediated signal. USP8 induces the deubiquitination of TRAF6, TAB2, TAK1, p62, and BECN1, which are pivotal roles for NF-κB activation and autophagy induction. Notably, the LIHC patient with low USP8 mRNA expression showed markedly shorter survival time, whereas there was no significant difference in the other 18-human cancers. Importantly, the TCGA data analysis on LIHC and transcriptome analysis on the USP8 knockout (USP8KO) SK-HEP-1 cells revealed a significant correlation between USP8 and TRAF6, TAB2, TAK1, p62, and BECN1, and enhanced NF-κB-dependent and autophagy-related cancer progression/metastasis-related genes in response to LPS stimulation. Furthermore, USP8KO SK-HEP-1 cells showed an increase in cancer migration and invasion by TLR4 stimulation, and a marked increase of tumorigenicity and metastasis in xenografted NSG mice. The results demonstrate that USP8 is negatively implicated in the LIHC progression through the regulation of TRAF6-mediated signal for the activation of NF-κB activation and autophagy induction. Our findings provide useful insight into the LIHC pathogenesis of cancer progression.

11.
Clin Immunol ; 139(3): 321-35, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21429805

RESUMO

In this study, we explore the possibility of human T cell development in the liver of humanized mice generated by intrahepatic injection of CD34(+) hCB cells into conditioned NOD/SCID/IL-2Rγ(null)(NSG) newborn mice. The intrahepatic injection of CD34(+) hCB cells led to effective reconstitution of human myeloid and lymphoid lineage cells. In contrast to the previously reported Rag2(-/-)γ(c)(-/-) humanized mice, interestingly, the thymus function of humanized NSG mice was markedly reduced in terms of its size and cell contents, whereas the livers of humanized NSG mice profoundly contained double-positive (DP), hCD4 and hCD8 single positive (SP), hCD34(+)hCD38(lo)hCD1a(-) (TSP), hCD34(+)hCD38(hi)hCD1a(-) (ETP), and hCD34(+)hCD38(+)hCD1a(+) (pre-T cells) cells. Furthermore, immunostaining of the liver revealed that human T cells were co-localized with hDCs. Taken together, our results demonstrate that the intrahepatic injection of hCD34(+) hCB cells can facilitate human T cell development in the livers of humanized NSG mice.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Sangue Fetal/imunologia , Hematopoese/imunologia , Subunidade gama Comum de Receptores de Interleucina/imunologia , Fígado/imunologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Citometria de Fluxo , Humanos , Imunofenotipagem , Fígado/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID
12.
J Clin Immunol ; 31(2): 253-64, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20981478

RESUMO

BACKGROUND: Busulfan treatment as a chemotherapeutic agent has been considered an alternative approach in xenograft model because it offers a simple, convenient, effective, and less toxic conditioning regimen. OBJECTIVE AND METHODS: To investigate busulfan effects on the reconstitution of human immune cells and the generation of immune response to foreign antigens, we generated humanized NOD/SCID/IL-2Rγ(null) (NSG) mice conditioned either busulfan or total body irradiation (TBI) with hCD34(+) CB cells. RESULTS: Busulfan resulted in a high survival rate and effective reconstitution of human immune cells including B, T, macrophage, and dendritic cells in humanized NSG mice, compared to that of TBI. Moreover, the humanized NSG mice conditioned busulfan showed effective B cell development and thereby the high production of human antibody against immunized antigen. CONCLUSION: Humanized mice conditioned by busulfan provide a powerful and versatile tool for studying the entire process of human B-lymphocyte development and for producing specific human antibodies.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Bussulfano/farmacologia , Hospedeiro Imunocomprometido/efeitos dos fármacos , Adjuvantes Imunológicos/farmacologia , Animais , Formação de Anticorpos/efeitos da radiação , Linfócitos B/efeitos dos fármacos , Linfócitos B/efeitos da radiação , Transplante de Células-Tronco Hematopoéticas , Humanos , Hospedeiro Imunocomprometido/efeitos da radiação , Subunidade gama Comum de Receptores de Interleucina/genética , Dose Letal Mediana , Linfonodos/citologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Análise de Sobrevida , Linfócitos T/imunologia , Irradiação Corporal Total
13.
J Clin Immunol ; 31(4): 699-709, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21544592

RESUMO

BACKGROUND: In terms of the function and reconstitution efficacy of human immune cells, co-transplantation of human fetal tissues, such as thymus and liver, with CD34(+) hematopoietic stem cells (HSCs) has potential advantages in the generation of humanized mice. OBJECTIVE AND METHODS: To examine the effects of bone tissues in the reconstitution of human immune cells, particularly in B cells, we generated a new humanized mice co-transplanted with human fetal thymus (hFT)/fetal bone (hFB) tissues and human fetal liver-derived CD34(+) cells. RESULTS: Humanized mice exhibited effective reconstitution of human immune cells earlier compared to control humanized mice. In terms of quantity, the number of immune cells, such as human T, B, and monocyte/macrophages was significantly increased. Furthermore, significant increase of B cell progenitors and immature/naïve B cells could be detected in the bone marrow and spleen of humanized mice. CONCLUSION: Our results demonstrate that co-transplantation of hFB tissue may facilitate the reconstitution of human B and T cells, and therefore the humanized model may be used to develop therapeutic human antibodies for clinical use.


Assuntos
Linfócitos B/imunologia , Transplante Ósseo , Osso e Ossos/imunologia , Transplante de Células-Tronco Hematopoéticas , Timo/transplante , Transplante Heterólogo/imunologia , Animais , Antígenos CD34/biossíntese , Linhagem da Célula/imunologia , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/imunologia , Fígado/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID
14.
Sci Rep ; 11(1): 10603, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-34011992

RESUMO

Human delta-like 1 (hDlk1) is known to be able to regulate cell fate decisions during hematopoiesis. Mesenchymal stromal cells (MSCs) are known to exhibit potent immunomodulatory roles in a variety of diseases. Herein, we investigated in vivo functions of hDlk1-hMSCs and hDlk1+hMSCs in T cell development and T cell response to viral infection in humanized NOD/SCID/IL-2Rγnull (NSG) mice. Co-injection of hDlk1-hMSC with hCD34+ cord blood (CB) cells into the liver of NSG mice markedly suppressed the development of human T cells. In contrast, co-injection of hDlk1+hMSC with hCD34+ CB cells into the liver of NSG dramatically promoted the development of human T cells. Human T cells developed in humanized NSG mice represent markedly diverse, functionally active, TCR V[Formula: see text] usages, and the restriction to human MHC molecules. Upon challenge with Epstein-Barr virus (EBV), EBV-specific hCD8+ T cells in humanized NSG mice were effectively mounted with phenotypically activated T cells presented as hCD45+hCD3+hCD8+hCD45RO+hHLA-DR+ T cells, suggesting that antigen-specific T cell response was induced in the humanized NSG mice. Taken together, our data suggest that the hDlk1-expressing MSCs can effectively promote the development of human T cells and immune response to exogenous antigen in humanized NSG mice. Thus, the humanized NSG model might have potential advantages for the development of therapeutics targeting infectious diseases in the future.


Assuntos
Antígenos/imunologia , Proteínas de Ligação ao Cálcio/metabolismo , Imunidade , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Antígenos CD34/metabolismo , Infecções por Vírus Epstein-Barr/imunologia , Sangue Fetal/citologia , Feto/embriologia , Herpesvirus Humano 4/imunologia , Humanos , Fígado/citologia , Fígado/embriologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos NOD , Camundongos SCID
15.
Nanomedicine ; 6(2): 263-76, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19699324

RESUMO

Live imaging is a powerful technique that can be used to characterize the fate and location of stem cells in animal models. Here we investigated the characteristics and in vitro cytotoxicity of human mesenchymal stem cells (MSCs) labeled with silica-coated magnetic nanoparticles incorporating rhodamine B isothiocyanate, MNPs@SiO2(RITC). We also conducted various in vivo-uptake tests with nanoparticle-labeled human MSCs. MNPs@SiO2(RITC) showed photostability against ultraviolet light exposure and were nontoxic to human MSCs, based on the MTT, apoptosis, and cell cycle arrest assays. In addition, MNPs@SiO2(RITC) did not affect the surface phenotype or morphology of human MSCs. We also demonstrated that MNPs@SiO2(RITC) have stable retention properties in MSCs in vitro. Furthermore, using optical and magnetic resonance imaging, we successfully detected a visible signal from labeled human MSCs that were transplanted into NOD.CB17-Prkdc(SCID) (NOD-SCID) mice. These results demonstrate that MNPs@SiO2(RITC) are biocompatible and useful tools for human MSC labeling and bioimaging. FROM THE CLINICAL EDITOR: The characteristics and in vitro cytotoxicity of human mesenchymal stem cells (MSCs) labeled with silica-coated magnetic nanoparticles incorporating rhodamine B isothiocyanate, RITC were investigated in this study. RITC showed photostability against ultraviolet light exposure and was nontoxic to human MSCs. Using both optical and magnetic resonance imaging, successful detection of signal from labeled human MSCs transplanted into mice is demonstrated.


Assuntos
Portadores de Fármacos/química , Sangue Fetal/citologia , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais/citologia , Nanopartículas/efeitos adversos , Rodaminas , Dióxido de Silício/química , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Meios de Contraste/química , Portadores de Fármacos/efeitos adversos , Sangue Fetal/efeitos dos fármacos , Humanos , Aumento da Imagem/métodos , Magnetismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Rodaminas/efeitos adversos , Coloração e Rotulagem/métodos
16.
Cancer Lett ; 478: 56-69, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32145342

RESUMO

The efficacy of an immune checkpoint blockade has been demonstrated against various types of cancer, but its suitability has not been fully proven for therapies specifically targeting sarcoma. We conducted a pan-cancer tumor data analysis to identify key immune-related variables strongly associated with sarcoma prognosis, and we explored whether these expected factors are functionally correlated with anti-PD-1 therapy in humanized (Hu) NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice xenografted with dedifferentiated liposarcoma (DDLPS). We found that an abundance of hCD8+ T cells and hNK cells was functionally associated with anti-PD-1 effects in the Hu-NSG DDLPS mice. Phenotypically, these cells were shown to be hCD8+IFNγ+, hCD8+PD-1+, hCD8+Ki-67+, hCD56+IFNγ+, hCD56+PD-1+, and hCD56+Ki-67+ cells and were enriched in splenocytes and tumor-infiltrating lymphocytes (TILs) of Hu-NSG DDLPS mice treated with anti-PD-1 antibody. Moreover, a considerable increase in activated hCD56+NKp46+NKG2D+ NK cells was also detected. Our findings suggest that hCD8+ T and hNK subsets play a pivotal role in anti-DDLPS tumor effects of anti-PD-1 therapy. The results provide clinical reference for advanced anti-PD-1 therapy targeting sarcoma tumors including DDLPS.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Linfócitos T CD8-Positivos/metabolismo , Células Matadoras Naturais/metabolismo , Lipossarcoma/tratamento farmacológico , Neoplasias Retroperitoneais/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Lipossarcoma/imunologia , Lipossarcoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Prognóstico , Neoplasias Retroperitoneais/imunologia , Neoplasias Retroperitoneais/patologia , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Transplantation ; 94(11): 1095-102, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23222735

RESUMO

BACKGROUND: In many humanized mouse models, there are few T cells in the engrafted human cell, whereas the number of B cells is high. We attempted to overcome this limitation and investigate whether the entire process of human T cell development arose similarly to the process in humans, as previously reported. METHODS: To produce an advanced humanized mice model, we transplanted human fetal liver/thymus tissue subrenally and injected human CD34(+) stem cells intravenously into NOD/SCID/IL2Rgamma null (NSG) mice. RESULTS: Humanized mice transplanted with fetal thymus/liver tissues and fetal liver-derived CD34(+) stem cells (FLT+FLCD34) showed higher levels of human cells and T cells than mice transplanted with fetal liver-derived CD34(+) stem cells only (FLCD34). In the transplanted thymus tissue of FLT+FLCD34 mice, thymus seeding progenitors (TSPs), early thymic progenitors (ETPs), pre-T cells, and all the other human T cell populations were identified. In the periphery, FLT+FLCD34 mice have high levels of CD45RA(+) T cells; conversely, FLCD34 mice have higher levels of CD45RO(+) T cells. The CD45RO(+) T cells of FLCD34 mice proliferated rapidly after stimulation and exhibited innate T cells properties, expressing PLZF (promyelocytic leukemia zinc finger protein). CONCLUSION: Human T cells educated by mouse MHC II in mice without a human thymus differ from normal human T cells. On the basis of these findings, numerous T cell-tropic human diseases could be explored in our humanized mice and molecular aspects of human T cell development could be also studied extensively.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Transplante de Fígado , Fígado , Linfócitos T/transplante , Timo/transplante , Animais , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Proliferação de Células , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Humanos , Imunidade Inata , Memória Imunológica , Imunofenotipagem/métodos , Interferon gama/metabolismo , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Fígado/embriologia , Fígado/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células Precursoras de Linfócitos T/imunologia , Células Precursoras de Linfócitos T/transplante , Proteína com Dedos de Zinco da Leucemia Promielocítica , Linfócitos T/imunologia , Timócitos/imunologia , Timócitos/transplante , Timo/embriologia , Timo/imunologia , Fatores de Tempo
19.
Transplantation ; 89(5): 509-17, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20125064

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs), also known as multipotent progenitor cells, release several factors that support cell survival and enhance wound healing. We hypothesized that MSC-secreted molecules would induce a trophic effect in pancreatic islet culture conditions. METHODS: Pancreatic islets were co-cultured with MSCs, and ADP/ATP ratios, glucose stimulated insulin secretion (GSIS), and DNA fragmentation were evaluated to measure islet quality and viability in vitro. The induction of signal molecules related to the control of survival, function, and angiogenesis was also analyzed. Cell quality assays, DNA fragmentation assays, and islet transplantation into streptozotocin-induced diabetic mice were performed using MSC-conditioned medium (CM)-cultured islets. Furthermore, we identified soluble molecules within MSC-CM. RESULTS: Islets co-cultured with MSCs demonstrated lower ADP/ATP ratios, and higher GSIS indexes and viability. Furthermore, co-cultured islets revealed higher levels of anti-apoptotic signal molecules (X-linked inhibitor of apoptosis protein, Bcl-xL, Bcl-2, and heat shock protein-32) and demonstrated increased vascular endothelial growth factor receptor 2 and Tie-2 mRNA expression and increased levels of phosphorylated Tie-2 and focal adhesion kinase protein. Islets cultured in MSC-CM demonstrated lower ADP/ATP ratios, less apoptosis, and a higher GSIS indexes. Diabetic mice that received islet transplants (200 islet equivalent) cultured in MSC-CM for 48 hr demonstrated significantly lower blood glucose levels and enhanced blood vessel formation. In addition, interleukin-6, interleukin-8, vascular endothelial growth factor-A, hepatocyte growth factor, and transforming growth factor-beta were detected at significant levels in MSC-CM. CONCLUSIONS: These results suggest that the trophic factors secreted by human MSCs enhance islet survival and function after transplantation.


Assuntos
Transplante das Ilhotas Pancreáticas/fisiologia , Células-Tronco Mesenquimais/citologia , Adenocarcinoma , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Linhagem Celular , Sobrevivência Celular , Técnicas de Cocultura , Neoplasias do Colo , Sangue Fetal/citologia , Teste de Tolerância a Glucose , Humanos , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/patologia , Masculino , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Células-Tronco/fisiologia , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA