Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Front Immunol ; 13: 853349, 2022.
Article in English | MEDLINE | ID: mdl-35757709

ABSTRACT

Islet transplantation to treat the late stage of type 1 diabetic patient (T1DM) has recently made inspiring success in clinical trials. However, most patients experience a decline in islet graft function in one to three years due to immune rejection. Although the mechanisms of immune cells, including macrophages, dendritic cells (DCs), neutrophils, natural killer cells (NKs), B cells, and T cells, that mediate immune rejection have been investigated, the overall characteristics of immune infiltrates in islet allografts and syngeneic grafts remain unclear. Single-cell RNA sequencing (scRNA-seq) has provided us with new opportunities to study the complexity of the immune microenvironment in islet transplants. In the present study, we used scRNA-seq to comprehensively analyze the immune heterogeneity in the mouse model of islet transplantation. Our data revealed T lymphocytes and myeloid cells as the main immune components of grafts 7 days post-islet transplantation, especially in allografts. Moreover, our results indicated that allogeneic islet cells were transformed into antigen-presenting cell-like cells with highly expressed MHC class I molecules and genes involved in MHC class I-mediated antigen presentation. This transformation may dramatically facilitate the interaction with cytotoxic CD8+ T cells and promote the destruction of islet allografts. Our study provides insight into the transcriptomics and diverse microenvironment of islet grafts and their impacts on immune rejection.


Subject(s)
CD8-Positive T-Lymphocytes , Islets of Langerhans Transplantation , Allografts , Animals , Histocompatibility Antigens Class I , Humans , Isografts , Mice , Transplantation, Homologous
2.
Transplantation ; 105(9): 1980-1988, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34416751

ABSTRACT

BACKGROUND: Type 1 diabetes (T1DM) is a chronic autoimmune disease characterized by T-cell-mediated destruction of insulin-producing beta cells. Evidence shows that patients with T1DM and mice used in specific diabetic models both exhibit changes in their intestinal microbiota and dysregulated microbiota contributes to the pathogenesis of T1DM. Islet transplantation (Tx) is poised to play an important role in the treatment of T1DM. However, whether treatment of T1DM with islet Tx can rescue dysregulated microbiota remains unclear. METHODS: In this study, we induced diabetic C57BL/6 mice with streptozotocin. Then treatment with either insulin administration, or homogenic or allogenic islet Tx was performed to the diabetic mice. Total DNA was isolated from fecal pellets and high-throughput 16S rRNA sequencing was used to investigate intestinal microbiota composition. RESULTS: The overall microbial diversity was comparable between control (nonstreptozotocin treated) and diabetic mice. Our results showed the ratio of the Bacteroidetes: Firmicutes between nondiabetic and diabetic mice was significant different. Treatment with islet Tx or insulin partially corrects the dysregulated bacterial composition. At the genus level, Bacteroides, Odoribacter, and Alistipes were associated with the progression and treatment efficacy of the disease, which may be used as a biomarker to predict curative effect of treatment for patients with T1DM. CONCLUSIONS: Collectively, our results indicate that diabetic mice show changed microbiota composition and that treatment with insulin and islet Tx can partially correct the dysregulated microbiota.


Subject(s)
Bacteria/growth & development , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Gastrointestinal Microbiome , Glycemic Control , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Animals , Bacteria/classification , Bacteria/genetics , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/microbiology , Dysbiosis , Feces/microbiology , Islets of Langerhans Transplantation , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Ribotyping , Streptozocin , Tissue Culture Techniques
3.
J Vis Exp ; (156)2020 02 16.
Article in English | MEDLINE | ID: mdl-32116299

ABSTRACT

Pancreatic islet transplantation is a well-established therapeutic treatment for type 1 diabetes. The kidney capsule is the most commonly used site for islet transplantation in rodent models. However, the tight kidney capsule limits the transplantation of sufficient islets in large animals and humans. The inguinal subcutaneous white adipose tissue (ISWAT), a new subcutaneous space, was found to be a potentially valuable site for islet transplantation. This site has better blood supply than other subcutaneous spaces. Moreover, the ISWAT accommodates a larger islet mass than the kidney capsule, and transplantation into it is simple. This manuscript describes the procedure of mouse islet isolation and transplantation in the ISWAT site of syngeneic diabetic mouse recipients. Using this protocol, murine pancreatic islets were isolated by standard collagenase digestion and a basement membrane matrix hydrogel was used for fixing the purified islets in the ISWAT site. The blood glucose levels of the recipient mice were monitored for more than 100 days. Islet grafts were retrieved at day 100 after transplantation for histological analysis. The protocol for islet transplantation in the ISWAT site described in this manuscript is simple and effective.


Subject(s)
Inguinal Canal/anatomy & histology , Islets of Langerhans Transplantation , Models, Biological , Subcutaneous Fat/transplantation , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/therapy , Graft Survival , Humans , Islets of Langerhans/pathology , Mice, Inbred C57BL , Perfusion , Tissue Survival
4.
Ophthalmic Res ; 63(3): 224-233, 2020.
Article in English | MEDLINE | ID: mdl-31962334

ABSTRACT

Controversial results regarding the associations between aldose reductase (AR) genetic polymorphisms and diabetic retinopathy (DR) have been reported for many years. The present meta-analysis was performed to clarify the effects of the AR gene C(-106)T polymorphism on DR risk. The PubMed, Web of Sciences, Cochrane library, EMBASE, Chinese National Knowledge Infrastructure, and Wan Fang databases were extensively searched in Chinese to select relevant studies with an updated date of April 25, 2018. The Newcastle-Ottawa Scale (NOS) was applied to assess quality. The random-effects model was applied to calculate the pooled OR and 95% CI. This meta-analysis identified 23 studies with an average score of 7.52 for NOS analysis, including 4,313 DR cases and 5,128 diabetes mellitus (DM) control cases. In the overall analysis, a significant association between the AR gene C(-106)T polymorphism and DR susceptibility was found. In subgroups stratified by DM type and ethnicity, significantly increased risks for DR were found in DM type 1, East Asian populations, and Middle Eastern populations. Compared with DR control cases, the following associations were found: T vs. C: OR 0.91, 95% CI 0.85-0.97, I2 = 72.9%; CT + TT vs. CC: OR 0.75, 95% CI 0.68-0.81, I2 = 86.7%; and CT vs. CC: OR 0.86, 95% CI 0.78-0.94, I2 = 70.5%. The results of this meta-analysis showed a significant association between the AR gene C(-106)T polymorphism and susceptibility to DR in DM patients. DM patients with allele T and CT+TT genotype of the AR gene may have a lower risk of DR.


Subject(s)
Aldehyde Reductase/genetics , DNA/genetics , Diabetic Retinopathy/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Aldehyde Reductase/metabolism , Alleles , Diabetic Retinopathy/metabolism , Gene Frequency , Genotype , Humans , Risk Factors
5.
Mol Cell ; 72(2): 380-394.e7, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30293782

ABSTRACT

RNA splicing is a critical mechanism by which to modify transcriptome, and its dysregulation is the underlying cause of many human diseases. It remains challenging, however, to genetically modulate a splicing event in its native context. Here, we demonstrate that a CRISPR-guided cytidine deaminase (i.e., targeted-AID mediated mutagenesis [TAM]) can efficiently modulate various forms of mRNA splicing. By converting invariant guanines to adenines at either 5' or 3' splice sites (SS), TAM induces exon skipping, activation of alternative SS, switching between mutually exclusive exons, or targeted intron retention. Conversely, TAM promotes downstream exon inclusion by mutating cytidines into thymines at the polypyrimidine tract. Applying this approach, we genetically restored the open reading frame and dystrophin function of a mutant DMD gene in patient-derived induced pluripotent stem cells (iPSCs). Thus, the CRISPR-guided cytidine deaminase provides a versatile genetic platform to modulate RNA splicing and to correct mutations associated with aberrant splicing in human diseases.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cytidine Deaminase/genetics , RNA Splicing/genetics , Amino Acid Sequence , Animals , Cell Line , Dystrophin/genetics , Exons/genetics , Gene Regulatory Networks , HEK293 Cells , Humans , Introns/genetics , Mice , Open Reading Frames/genetics , RNA Splice Sites/genetics
6.
J Biol Chem ; 292(29): 12285-12295, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28611064

ABSTRACT

Increasing evidence indicates that alternative processing of mRNA, including alternative splicing, 3' alternative polyadenylation, and regulation of mRNA stability/translation, represents a major mechanism contributing to protein diversification. For example, in alternative polyadenylation, the 3' end of the immunoglobulin heavy chain mRNA is processed during B cell differentiation, and this processing involves RNA-binding proteins. hnRNPLL (heterogeneous nuclear ribonucleoprotein L-like protein) is an RNA-binding protein expressed in terminally differentiated lymphocytes, such as memory T cells and plasma cells. hnRNPLL regulates various processes of RNA metabolism, including alternative pre-mRNA splicing and RNA stability. In plasma cells, hnRNPLL also regulates the transition from the membrane isoform of the immunoglobulin heavy-chain (mIgH) to the secreted isoform (sIgH), but the precise mechanism remains to be identified. In this study, we report that hnRNPLL specifically associates with cytoplasmic PABPC1 (poly(A)-binding protein 1) in both T cells and plasma cells. We found that although PABPC1 is not required for the alternative splicing of CD45, a primary target of hnRNPLL in lymphocytes, PABPC1 does promote the binding of hnRNPLL to the immunoglobulin mRNA and regulates switching from mIgH to sIgH in plasma cells. Given the recently identified role of PABPC1 in mRNA alternative polyadenylation, our findings suggest that PABPC1 recruits hnRNPLL to the 3'-end of RNA and regulates the transition from membrane Ig to secreted Ig through mRNA alternative polyadenylation. In conclusion, our study has revealed a mechanism that regulates immunoglobulin secretion in B cells via cooperation between a plasma cell-specific RBP (hnRNPLL) and a universally expressed RBP (PABPC1).


Subject(s)
Cytoplasm/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Immunoglobulin Heavy Chains/metabolism , Plasma Cells/metabolism , Poly(A)-Binding Protein I/metabolism , Polyadenylation , RNA, Messenger/metabolism , Animals , Cells, Cultured , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Immunoglobulin Class Switching , Immunoglobulin Heavy Chains/genetics , Immunoprecipitation , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Plasma Cells/cytology , Plasma Cells/immunology , Poly(A)-Binding Protein I/antagonists & inhibitors , Poly(A)-Binding Protein I/chemistry , Poly(A)-Binding Protein I/genetics , Protein Interaction Domains and Motifs , RNA Interference , Spleen/cytology , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Transplantation ; 99(9): 1796-806, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25919768

ABSTRACT

BACKGROUND: T cell-mediated immune rejection is a key barrier to islet transplantation. Preliminary studies have shown that arsenic trioxide (As2O3) can inhibit T cell responses and prolong heart allograft survival. Here, we sought to investigate the possibility of using As2O3 to prolong islet allograft survival in an acute rejection model of Balb/c to C57B/6 mice. METHODS: Recipient mice were treated with As2O3 and/or rapamycin after islet allograft transplantation. At day 10 after transplantation, the graft, spleen, lymph nodes, and blood of the recipient mice were recovered for analysis. In vitro, to further examine the mechanism underlying As2O3 protection of islet allografts against T cell-mediated rejection, mixed lymphocyte reaction and apoptosis analyses of T cells were performed. The phosphorylation levels of IκBα and p38 were also evaluated to confirm the proliferation and apoptosis of As2O3-treated T cells. RESULTS: We found that As2O3 prolonged islet allograft survival by reducing inflammatory reactions, influencing cytokine synthesis and secretion and T-cell subset proportions, and inhibiting T-cell responses. Furthermore, As2O3 and rapamycin showed a synergistic effect in suppressing islet allotransplant rejection. CONCLUSIONS: Arsenic trioxide may prevent allograft rejection by inhibiting T-cell proliferation and inducing T-cell apoptosis.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Graft Rejection/prevention & control , Graft Survival/drug effects , Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation , Islets of Langerhans/drug effects , Islets of Langerhans/surgery , Oxides/pharmacology , T-Lymphocytes/drug effects , Animals , Arsenic Trioxide , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , I-kappa B Proteins/metabolism , Immunity, Cellular/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/adverse effects , Lymphocyte Activation/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Phosphorylation , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Diabetes ; 63(10): 3458-69, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24834979

ABSTRACT

Islet transplantation is a therapeutic option for type 1 diabetes, but its long-term success is limited by islet allograft survival. Many factors imperil islet survival, especially the adverse effects and toxicity due to clinical immunosuppressants. Compound (Cpd) K is a synthesized analog of highly unsaturated fatty acids from Isatis tinctoria L. (Cruciferae). Here we investigated the therapeutic effect of Cpd K in diabetic mice and found that it significantly prolonged islet allograft survival with minimal adverse effects after 10 days. Furthermore, it reduced the proportion of CD4(+) and CD8(+) T cells in spleen and lymph nodes, inhibited inflammatory cell infiltration in allografts, suppressed serum interleukin-2 and interferon-γ secretion, and increased transforming growth factor-ß and Foxp3 mRNA expression. Surprisingly, Cpd K and rapamycin had a synergistic effect. Cpd K suppressed proliferation of naïve T cells by inducing T-cell anergy and promoting the generation of regulatory T cells. In addition, nuclear factor-κB signaling was also blocked. Taken together, these findings indicate that Cpd K may have a potential immunosuppressant effect on islet transplantation.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Ginsenosides/therapeutic use , Graft Survival/drug effects , Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Ginsenosides/pharmacology , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Interferon-gamma/blood , Interleukin-2/blood , Mice , Spleen/drug effects , Spleen/pathology
9.
Xenotransplantation ; 18(4): 252-61, 2011.
Article in English | MEDLINE | ID: mdl-21848543

ABSTRACT

BACKGROUND: Memory T cells are a significant barrier to the induction of transplant tolerance. Our previous study demonstrated that multiple applications of anti-CD44 monoclonal antibody (mAb) could significantly inhibit CD4(+) memory T cells from mediating rejection of cardiac allografts. Now, we sought to explore the effect and mechanism of anti-CD44 mAb on the rejection of islet allografts and xenografts mediated by CD4(+) memory T cells. METHODS: In this study, we first engrafted skin grafts of C57BL/6 (B6) mice or Dark Agouti (DA) rats onto BALB/c mice to induce donor-reactive memory T cells. We adoptively transferred purified CD4(+) memory T cells to BALB/c origin nude mice and then transplanted islet allografts and xenografts to produce the Allo-Tx and Xeno-Tx models, respectively. We subsequently administered multiple anti-CD44 mAb and observed changes in the survival times of the islet grafts. RESULTS: In the Allo-Tx model, the mean survival time (MST) of the grafts was 7.7 days in the isotype group, and 20.3 days in the anti-CD44 group. In the Xeno-Tx model, the MST of the grafts was 7.2 days in the isotype group and 8.2 days in the anti-CD44 group. Compared with the isotype group, CD4(+) T cells on the grafts in the anti-CD44 group were significantly decreased in both the Allo-Tx and Xeno-Tx models, but the proportion of CD4(+) memory T cells in the spleens and draining lymph nodes of the recipient nude mice in the anti-CD44 group was significantly decreased in the Allo-Tx model, while it was increased in the Xeno-Tx model. The production of donor-specific IgG antibody in the anti-CD44 group did not vary in the Allo-Tx model, while it was markedly elevated in the Xeno-Tx model. Furthermore, the expression of interferon gamma in the anti-CD44 group was markedly decreased in both the Allo-Tx and Xeno-Tx models, while the expression of IL-4 in the anti-CD44 group was significantly increased only in the Xeno-Tx model. CONCLUSION: Multiple applications of the anti-CD44 mAb could significantly inhibit donor-reactive CD4(+) memory T cells from rejecting grafts via a Th1-dependent pathway, but xenoreactive CD4(+) memory T cells can avoid the effects of anti-CD44 mAb to reject islet xenografts via a Th2-dependent pathway.


Subject(s)
Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/immunology , Hyaluronan Receptors/immunology , Islets of Langerhans Transplantation/immunology , Th2 Cells/immunology , Transplantation, Heterologous/immunology , Transplantation, Homologous/immunology , Animals , CD4 Antigens/immunology , Cell Proliferation , Diabetes Mellitus, Experimental , Female , Graft Survival/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Rats , Transplantation, Heterologous/pathology , Transplantation, Homologous/pathology
10.
Immunol Lett ; 138(2): 122-8, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21477620

ABSTRACT

Donor-reactive memory T cells mediated accelerated rejection is known as a barrier to the survival of transplanted organs. We investigated the combination of different monoclonal antibodies (mAbs) and donor-specific transfusion (DST) in memory T cells-based adoptive mice model. In the presence of donor-reactive memory T cells, the mean survival time (MST) of grafts in the anti-CD40L/LFA-1/DST group was 49.8d. Adding anti-CD44/CD70 mAbs to anti-CD40L/LFA-1/DST treatment. The MST was more than 100 d (MST>100 d). Compared with anti-CD40L/LFA-1/DST group, anti-CD40L/LFA-1/CD44/CD70/DST group notably reduced the expansion of memory T cells, enhanced the proportion of CD4+Foxp3+ regulatory T cells (Tregs) and suppressed donor-specific responses. Our data suggest that anti-CD40L/LFA-1/CD44/CD70mAbs and DST can synergistically inhibit accelerated rejection mediated by memory T cells to induce long-lived heart allograft acceptance in mice.


Subject(s)
Antibodies, Monoclonal , Graft Rejection , Graft Survival/immunology , Heart Transplantation , Immunomodulation , Isoantigens/immunology , Monocytes , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/immunology , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/immunology , Cell Proliferation/drug effects , Female , Graft Rejection/immunology , Graft Rejection/prevention & control , Heart Transplantation/immunology , Hyaluronan Receptors/immunology , Immunologic Memory , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/transplantation , Skin Transplantation/immunology , Spleen/cytology , Spleen/immunology , Transplantation, Homologous
11.
Transpl Immunol ; 24(4): 195-202, 2011 May.
Article in English | MEDLINE | ID: mdl-21396447

ABSTRACT

BACKGROUND: Alloreactive memory T cells are a major obstacle to transplantation acceptance due to their capacity for accelerated rejection. METHODS: C57BL/6 mice that had rejected BALB/c skin grafts 4 weeks earlier were used as recipients. The recipient mice were treated with anti-CD154/LFA-1 with or without anti-CD70 during the primary skin transplantation and anti-CD154/LFA-1 or not during the secondary transplantation of BALB/c heart. We evaluated the impact of combinations of antibody-mediated blockade on the generation of memory T cells and graft survival after fully MHC-mismatched transplantations. RESULTS: One month after the primary skin transplantation, the proportions of CD4(+) memory T cells/CD4(+) T cells and CD8(+)memory T cells/CD8(+) T cells in the anti-CD154/LFA-1 combination group were 47.32±4.28% and 23.18±2.77%, respectively. In the group that included anti-CD70 treatment, the proportions were reduced to 34.10±2.71% and 12.19±3.52% (P<0.05 when comparing the proportion of memory T cells between the two groups). The addition of anti-CD70 to the treatment regimen prolonged the mean survival time following secondary heart transplantation from 10days to more than 90days (P<0.001). Furthermore, allogenic proliferation of recipient splenic T cells and graft-infiltrating lymphocytes were significantly decreased. Meanwhile, the proportion of regulatory T cells was increased to 9.46±1.48% on day 100 post-transplantation (P<0.05). CONCLUSIONS: The addition of anti-CD70 to the anti-CD154/LFA-1 combination given during the primary transplantation reduced the generation of memory T cells. This therapy regimen provided a potential means to alleviate the accelerated rejection mediated by memory T cells during secondary heart transplantation and markedly prolong the survival of heart allografts.


Subject(s)
Graft Rejection/drug therapy , Heart Transplantation , Immunotherapy , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , CD27 Ligand/immunology , CD40 Ligand/immunology , Cells, Cultured , Graft Rejection/immunology , Humans , Immunization , Immunologic Memory , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transplantation Tolerance
12.
Xenotransplantation ; 17(6): 460-8, 2010.
Article in English | MEDLINE | ID: mdl-21158947

ABSTRACT

BACKGROUND: Donor-reactive memory T cells are known to accelerate allograft rejection; in our previous study, we reported that combined monoclonal antibodies (mAbs) could prolong islet allograft survival in alloantigen-primed mice. In this study, we examine the effects of donor-reactive memory T cells on the xenograft survival and methods to prolong the islet graft survival. METHODS: To collect donor-reactive T cells, we performed full-thickness rat skin xenografting on BALB/c mice and isolated the T cells from the mice after 6-8 weeks. These cells were then adoptively transferred to syngenic mice 1 day before rat-to-mouse islet transplantation. Three experimental groups were established in the adoptive transfer model: recipient mice treated with isotype mAbs (isotype group); mice treated with anti-CD40L mAb (anti-CD40L group); and mice treated with anti-CD40L, anti-OX40L, and anti-CD122 mAbs (3-combined group). RESULTS: Lewis rat islet xenografts transplanted in naïve mice showed a mean survival time (MST) of 12.8 days, while the graft rejection was accelerated if the recipient mice were treated with adoptively transferred donor-reactive T cells (MST, 8.67 days). Treatment with anti-CD40L mb could not reverse the accelerated rejection (MST, 9.3 days). However, when anti-CD40L mb was combined with anti-OX40L and anti-CD122 mAbs, there was a considerable increase in the MST, which was 72.2 days. Compared to the isotype group, the 3-combined group had significantly lesser proportion of memory T cells and greater proportion of regulatory T cells (Tregs) in the spleen. Meanwhile, in the 3-combined group, the production of anti-rat antibodies was markedly inhibited. CONCLUSION: Treatment with a combination of antibodies could significantly reverse the accelerated rejection mediated by donor-reactive memory T cells by inhibiting cellular and humoral immune responses.


Subject(s)
Antibodies/immunology , Antigens, Heterophile/immunology , Graft Rejection/immunology , Immunologic Memory/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Islets of Langerhans Transplantation/immunology , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Inbred Lew
13.
Biochem Biophys Res Commun ; 341(3): 804-9, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16442077

ABSTRACT

CD38 has multiple roles in biology, including T lymphocyte signaling, neutrophil migration, neurotransmission, cell proliferation, apoptosis, and bone remodeling. To study the transcriptional control of the CD38 gene, we cloned a putative 1.8 kb promoter fragment from a rabbit genomic DNA library. Primer extension analysis indicated two transcription start sites consistent with the absence of a TATA box. Sequence analysis revealed several AP-1, AP-4, myo-D, GATA, and SP-1 sequences. MC3T3.E1 (osteoblast) or RAW-C3 (osteoclast precursor macrophage) cells were then transfected with the CD38 promoter or its deletion fragments ligated to the luciferase reporter gene. Except for the shortest 41 bp fragment, all fragments showed significant luciferase activity. There was a marked stimulation of basal activity in the 93 bp fragment that contained a GC box and SP-1 site. Furthermore, there were significant differences in the activity of the fragments in MC3T3.E1 and RAW-C3 cells. Intracellular Ca(2+) elevations by ionomycin (10muM) in MC3T3.E1 cells inhibited promoter activity, except in the short 41 bp. In contrast, cAMP elevation by exposure to forskolin (100 microM) inhibited activation of all fragments, except the 0.6 and 1.2kb fragments. Finally, TNF-alpha stimulated promoter activity in RAW-C3 cells transfected with the 93 bp and 1.0 kb fragments, consistent with the stimulation of CD38 mRNA by TNF-alpha. Physiologically, therefore, modulation of the expression of the NAD(+)-sensing enzyme, CD38, by Ca(2+), cAMP, and cytokines, such as TNF-alpha may contribute to coupling the intense metabolic activity of osteoclasts and osteoblasts to their respective bone-resorbing and bone-forming functions.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Gene Expression Regulation/genetics , Promoter Regions, Genetic/genetics , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Mice , Molecular Sequence Data , Osteoclasts/metabolism , Rabbits , Transcription, Genetic/genetics , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...