Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 162
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Transplant ; 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38219866

ABSTRACT

Mouse models have been instrumental in understanding mechanisms of transplant rejection and tolerance, but cross-study reproducibility and translation of experimental findings into effective clinical therapies are issues of concern. The Mouse Models in Transplantation symposium gathered scientists and physician-scientists involved in basic and clinical research in transplantation to discuss the strengths and limitations of mouse transplant models and strategies to enhance their utility. Participants recognized that increased procedure standardization, including the use of prespecified, defined endpoints, and statistical power analyses, would benefit the field. They also discussed the generation of new models that incorporate environmental and genetic variables affecting clinical outcomes as potentially important. If implemented, these strategies are expected to improve the reproducibility of mouse studies and increase their translation to clinical trials and, ideally, new Food and Drug Administration-approved drugs.

2.
Am J Transplant ; 22(3): 745-760, 2022 03.
Article in English | MEDLINE | ID: mdl-34704345

ABSTRACT

A safe, efficacious, and clinically applicable immunosuppressive regimen is necessary for islet xenotransplantation to become a viable treatment option for diabetes. We performed intraportal transplants of wild-type adult porcine islets in 25 streptozotocin-diabetic cynomolgus monkeys. Islet engraftment was good in 21, partial in 3, and poor in 1 recipient. Median xenograft survival was 25 days with rapamycin and CTLA4Ig immunosuppression. Adding basiliximab induction and maintenance tacrolimus to the base regimen significantly extended median graft survival to 147 days (p < .0001), with three animals maintaining insulin-free xenograft survival for 265, 282, and 288 days. We demonstrate that this regimen suppresses non-Gal anti-pig antibody responses, circulating effector memory T cell expansion, effector function, and infiltration of the graft. However, a chronic systemic inflammatory state manifested in the majority of recipients with long-term graft survival indicated by increased neutrophil to lymphocyte ratio, IL-6, MCP-1, CD40, and CRP expression. This suggests that this immunosuppression regimen fails to regulate innate immunity and resulting inflammation is significantly associated with increased incidence and severity of adverse events making this regimen unacceptable for translation. Additional studies are needed to optimize a maintenance regimen for regulating the innate inflammatory response.


Subject(s)
Diabetes Mellitus , Islets of Langerhans Transplantation , Animals , Graft Rejection/etiology , Graft Survival , Heterografts , Humans , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Inflammation/etiology , Islets of Langerhans Transplantation/methods , Macaca fascicularis , Swine , Transplantation, Heterologous/methods
3.
Am J Respir Crit Care Med ; 204(9): 1060-1074, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34346860

ABSTRACT

Rationale: Primary graft dysfunction (PGD) is a severe form of acute lung injury, leading to increased early morbidity and mortality after lung transplant. Obesity is a major health problem, and recipient obesity is one of the most significant risk factors for developing PGD. Objectives: We hypothesized that T-regulatory cells (Tregs) are able to dampen early ischemia-reperfusion events and thereby decrease the risk of PGD, whereas that action is impaired in obese recipients. Methods: We evaluated Tregs, T cells, and inflammatory markers, plus clinical data, in 79 lung transplant recipients and 41 liver or kidney transplant recipients and studied two groups of mice on a high-fat diet (HFD), which did ("inflammatory" HFD) or did not ("healthy" HFD) develop low-grade inflammation with decreased Treg function. Measurements and Main Results: We identified increased levels of IL-18 as a previously unrecognized mechanism that impairs Tregs' suppressive function in obese individuals. IL-18 decreases levels of FOXP3, the key Treg transcription factor, decreases FOXP3 di- and oligomerization, and increases the ubiquitination and proteasomal degradation of FOXP3. IL-18-treated Tregs or Tregs from obese mice fail to control PGD, whereas IL-18 inhibition ameliorates lung inflammation. The IL-18-driven impairment in Tregs' suppressive function before transplant was associated with an increased risk and severity of PGD in clinical lung transplant recipients. Conclusions: Obesity-related IL-18 induces Treg dysfunction that may contribute to the pathogenesis of PGD. Evaluation of Tregs' suppressive function together with evaluation of IL-18 levels may serve as a screening tool to identify obese individuals with an increased risk of PGD before transplant.


Subject(s)
Acute Lung Injury/etiology , Interleukin-18/metabolism , Lung Transplantation/adverse effects , Obesity/complications , Primary Graft Dysfunction/etiology , Reperfusion Injury/etiology , T-Lymphocytes, Regulatory/metabolism , Acute Lung Injury/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Mice, Obese , Middle Aged , Primary Graft Dysfunction/physiopathology , Reperfusion Injury/physiopathology
4.
Metabolomics ; 16(5): 65, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32367163

ABSTRACT

To examine metabolic differences between renal allograft acute cellular rejection (ACR) and ischemic-reperfusion injury (IRI), we transplanted MHC-mismatched kidneys and induced 28 min warm-IRI, and collected the ACR and IRI kidneys as well as their respective native and collateral control kidneys. We extracted metabolites from the kidney tissues and found the lysine catabolite saccharopine 12.5-fold enriched in IRI kidneys, as well as the immunometabolites itaconate and kynurenine in ACR kidneys. Saccharopine accumulation is known to be toxic to mitochondria and may contribute to IRI pathophysiology, while itaconate and kynurenine may be reflective of counterregulatory responses to immune activation in ACR.


Subject(s)
Graft Rejection/metabolism , Kidney/metabolism , Kynurenine/metabolism , Lysine/analogs & derivatives , Reperfusion Injury/metabolism , Succinates/metabolism , Animals , Disease Models, Animal , Female , Kidney/injuries , Kidney Transplantation/adverse effects , Lysine/metabolism , Metabolomics , Mice , Mice, Inbred C57BL
5.
FASEB J ; 33(1): 1098-1109, 2019 01.
Article in English | MEDLINE | ID: mdl-30102568

ABSTRACT

Alterations in gut microbiota are known to affect intestinal inflammation and obesity. Antibiotic treatment can affect weight gain by elimination of histone deacetylase (HDAC) inhibitor-producing microbes, which are anti-inflammatory by augmenting regulatory T (Treg) cells. We asked whether mice that lack HDAC6 and have potent suppressive Treg cells are protected from microbiota-induced accelerated weight gain. We crossed wild-type and HDAC6-deficient mice and subjected the offspring to perinatal penicillin, inducing weight gain via microbiota disturbance. We observed that male HDAC6-deficient mice were not protected and developed profoundly accelerated weight gain. The antibiotic-exposed HDAC6-deficient mice showed a mixed immune phenotype with increased CD4+ and CD8+ T-cell activation yet maintained the enhanced Treg cell-suppressive function phenotype characteristic of HDAC6-deficient mice. 16S rRNA sequencing of mouse fecal samples reveals that their microbiota diverged with time, with HDAC6 deletion altering microbiome composition. On a high-fat diet, HDAC6-deficient mice were depleted in representatives of the S24-7 family and Lactobacillus but enriched with Bacteroides and Parabacteroides; these changes are associated with obesity. Our findings further our understanding of the influence of HDACs on microbiome composition and are important for the development of HDAC6 inhibitors in the treatment of human diseases.-Lieber, A. D., Beier, U. H., Xiao, H., Wilkins, B. J., Jiao, J., Li, X. S., Schugar, R. C., Strauch, C. M., Wang, Z., Brown, J. M., Hazen, S. L., Bokulich, N. A., Ruggles, K. V., Akimova, T., Hancock, W. W., Blaser, M. J. Loss of HDAC6 alters gut microbiota and worsens obesity.


Subject(s)
Gastrointestinal Microbiome , Histone Deacetylase 6/physiology , Obesity/genetics , Obesity/microbiology , Animals , Bacteroides/isolation & purification , Diet, High-Fat , Fatty Liver/genetics , Feces , Germ-Free Life , Histone Deacetylase 6/genetics , Hyperlipidemias/genetics , Lactobacillus/isolation & purification , Male , Mesentery/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/immunology , Up-Regulation , Weight Gain
6.
Blood ; 130(2): 146-155, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28550044

ABSTRACT

Histone acetylation and the families of enzymes responsible for controlling these epigenetic marks have been implicated in regulating T-cell maturation and phenotype. Here, we demonstrate a previously undefined role of histone deacetylase 11 (HDAC11) in regulating T-cell effector functions. Using EGFP-HDAC11 transgenic reporter mice, we found that HDAC11 expression was lower in effector relative to naive and central memory T-cell populations, and activation of resting T cells resulted in its decreased expression. Experiments using HDAC11 knockout (KO) mice revealed that T cells from these mice displayed enhanced proliferation, proinflammatory cytokine production, and effector molecule expression. In addition, HDAC11KO T cells had increased expression of Eomesodermin (Eomes) and TBX21 (Tbet), transcription factors previously shown to regulate inflammatory cytokine and effector molecule production. Conversely, overexpression of HDAC11 resulted in decreased expression of these genes. Chromatin immunoprecipitation showed the presence of HDAC11 at the Eomes and Tbet gene promoters in resting T cells, where it rapidly disassociated following T-cell activation. In vivo, HDAC11KO T cells were refractory to tolerance induction. HDAC11KO T cells also mediated accelerated onset of acute graft-versus-host disease (GVHD) in a murine model, characterized by increased proliferation of T cells and expression of interferon-γ, tumor necrosis factor, and EOMES. In addition, adoptive transfer of HDAC11KO T cells resulted in significantly reduced tumor burden in a murine B-cell lymphoma model. Taken together, these data demonstrate a previously unknown role of HDAC11 as a negative epigenetic regulator of T-cell effector phenotype and function.


Subject(s)
Gene Expression Regulation, Neoplastic , Graft vs Host Disease/immunology , Histone Deacetylase 1/genetics , Lymphoma, B-Cell/immunology , T-Box Domain Proteins/genetics , T-Lymphocytes/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Histone Deacetylase 1/deficiency , Histone Deacetylase 1/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mice , Mice, Knockout , Promoter Regions, Genetic , Signal Transduction , T-Box Domain Proteins/immunology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Int J Cancer ; 138(10): 2477-86, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26704363

ABSTRACT

Histone/protein deacetylases (HDACs) are frequently upregulated in human malignancies and have therefore become therapeutic targets in cancer therapy. However, inhibiting certain HDAC isoforms can have protolerogenic effects on the immune system, which could make it easier for tumor cells to evade the host immune system. Therefore, a better understanding of how each HDAC isoform affects immune biology is needed to develop targeted cancer therapy. Here, we studied the immune phenotype of HDAC5(-/-) mice on a C57BL/6 background. While HDAC5(-/-) mice replicate at expected Mendelian ratios and do not develop overt autoimmune disease, their T-regulatory (Treg) cells show reduced suppressive function in vitro and in vivo. Likewise, CD4(+) T-cells lacking HDAC5 convert poorly to Tregs under appropriately polarizing conditions. To test if this attenuated Treg formation and suppressive function translated into improved anticancer immunity, we inoculated HDAC5(-/-) mice and littermate controls with a lung adenocarcinoma cell line. Cumulatively, lack of HDAC5 did not lead to better anticancer immunity. We found that CD8(+) T cells missing HDAC5 had a reduced ability to produce the cytokine, IFN-γ, in vitro and in vivo, which may offset the benefit of weakened Treg function and formation. Taken together, targeting HDAC5 weakens suppressive function and de-novo induction of Tregs, but also reduces the ability of CD8(+) T cells to produce IFN-γ.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Forkhead Transcription Factors/metabolism , Gene Deletion , Gene Targeting , Interferon-gamma/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
8.
Kidney Int ; 89(5): 1016-1026, 2016 May.
Article in English | MEDLINE | ID: mdl-27083279

ABSTRACT

Current immunosuppressive medications used after transplantation have significant toxicities. Foxp3(+) T-regulatory cells can prevent allograft rejection without compromising protective host immunity. Interestingly, inhibiting the class III histone/protein deacetylase Sirtuin-1 can augment Foxp3(+) T-regulatory suppressive function through increasing Foxp3 acetylation. Here we determined whether Sirtuin-1 targeting can stabilize biological allograft function. BALB/c kidney allografts were transplanted into C57BL/6 recipients with a CD4-conditional deletion of Sirtuin-1 (Sirt1(fl/fl)CD4(cre)) or mice treated with a Sirtuin-1-specific inhibitor (EX-527), and the native kidneys removed. Blood chemistries and hematocrit were followed weekly. Sirt1(fl/fl)CD4(cre) recipients showed markedly longer survival and improved kidney function. Sirt1(fl/fl)CD4(cre) recipients exhibited donor-specific tolerance, accepted BALB/c, but rejected third-party C3H cardiac allografts. C57BL/6 recipients of BALB/c renal allografts that were treated with EX-527 showed improved survival and renal function at 1, but not 10 mg/kg/day. Pharmacologic inhibition of Sirtuin-1 also improved renal allograft survival and function with dosing effects having relevance to outcome. Thus, inhibiting Sirtuin-1 can be a useful asset in controlling T-cell-mediated rejection. However, effects on non-T cells that could adversely affect allograft survival and function merit consideration.


Subject(s)
Carbazoles/pharmacology , Graft Rejection/prevention & control , Graft Survival/drug effects , Histone Deacetylase Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Kidney Transplantation/adverse effects , Kidney/drug effects , Sirtuin 1/antagonists & inhibitors , Allografts , Animals , Female , Graft Rejection/enzymology , Graft Rejection/immunology , Graft Rejection/physiopathology , Kidney/enzymology , Kidney/immunology , Kidney/physiopathology , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Sirtuin 1/deficiency , Sirtuin 1/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Time Factors , Transplantation Tolerance/drug effects
9.
FASEB J ; 29(6): 2315-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25681462

ABSTRACT

Conventional T (Tcon) cells and Foxp3(+) T-regulatory (Treg) cells are thought to have differing metabolic requirements, but little is known of mitochondrial functions within these cell populations in vivo. In murine studies, we found that activation of both Tcon and Treg cells led to myocyte enhancer factor 2 (Mef2)-induced expression of genes important to oxidative phosphorylation (OXPHOS). Inhibition of OXPHOS impaired both Tcon and Treg cell function compared to wild-type cells but disproportionally affected Treg cells. Deletion of Pgc1α or Sirt3, which are key regulators of OXPHOS, abrogated Treg-dependent suppressive function and impaired allograft survival. Mef2 is inhibited by histone/protein deacetylase-9 (Hdac9), and Hdac9 deletion increased Treg suppressive function. Hdac9(-/-) Treg showed increased expression of Pgc1α and Sirt3, and improved mitochondrial respiration, compared to wild-type Treg cells. Our data show that key OXPHOS regulators are required for optimal Treg function and Treg-dependent allograft acceptance. These findings provide a novel approach to increase Treg function and give insights into the fundamental mechanisms by which mitochondrial energy metabolism regulates immune cell functions in vivo.


Subject(s)
Energy Metabolism/immunology , Forkhead Transcription Factors/immunology , Graft Survival/immunology , Mitochondria/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Blotting, Western , Energy Metabolism/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Graft Survival/genetics , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , MEF2 Transcription Factors/immunology , MEF2 Transcription Factors/metabolism , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Repressor Proteins/immunology , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 3/genetics , Sirtuin 3/immunology , Sirtuin 3/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
10.
Blood ; 121(18): 3631-9, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23444399

ABSTRACT

Protocols to use Foxp3+ T-regulatory (Treg) cells for cellular therapy, especially postallogeneic stem cell transplantation, are currently being developed and tested by various groups. Inhibitors of DNA methyltransferase (Dnmt) enzymes have been advocated as a means to promote and stabilize Foxp3 expression in Tregs undergoing expansion in vitro before their injection in vivo. We investigated the effects of conditionally deleting two Dnmt enzymes that co-immunoprecipitated with Foxp3 in Treg isolates. Deletion of Dnmt1, but not Dnmt3a, decreased the numbers and function of peripheral Tregs and impaired conversion of conventional T cells into Foxp3+ Tregs under polarizing conditions. Importantly, mice with conditional deletion of Dnmt1 in their Tregs died of autoimmunity by 3 to 4 weeks of age unless they were rescued by perinatal transfer of wild-type Tregs. Conditional Dnmt1 deletion did not affect methylation of CpG sites within Foxp3 but decreased global DNA methylation and altered Treg expression of several hundred pro-inflammatory and other genes. Hence, Dnmt1 is necessary for maintenance of the core gene program underlying Treg development and function, and its deletion within the Treg lineage leads to lethal autoimmunity. These data suggest that caution may be warranted when considering the use of DNMT inhibitors in development of Treg-based cellular therapies.


Subject(s)
Autoimmune Diseases/mortality , Autoimmune Diseases/prevention & control , DNA (Cytosine-5-)-Methyltransferases/genetics , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/physiology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/genetics , Autoimmunity/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/physiology , Gene Deletion , Gene Expression Regulation, Enzymologic/physiology , Genetic Therapy , Immunotherapy, Adoptive/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
11.
Am J Respir Crit Care Med ; 189(5): 567-75, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24467603

ABSTRACT

RATIONALE: Biologic pathways with significant genetic conservation across human populations have been implicated in the pathogenesis of primary graft dysfunction (PGD). The evaluation of the role of recipient genetic variation in PGD has thus far been limited to single, candidate gene analyses. OBJECTIVES: We sought to identify genetic variants in lung transplant recipients that are responsible for increased risk of PGD using a two-phase large-scale genotyping approach. METHODS: Phase 1 was a large-scale candidate gene association study of the multicenter, prospective Lung Transplant Outcomes Group cohort. Phase 2 included functional evaluation of selected variants and a bioinformatics screening of variants identified in phase 1. MEASUREMENTS AND MAIN RESULTS: After genetic data quality control, 680 lung transplant recipients were included in the analysis. In phase 1, a total of 17 variants were significantly associated with PGD, four of which were in the prostaglandin E2 family of genes. Among these were a coding variant in the gene encoding prostaglandin E2 synthase (PTGES2; P = 9.3 × 10(-5)) resulting in an arginine to histidine substitution at amino acid position 298, and three variants in a block containing the 5' promoter and first intron of the PTGER4 gene (encoding prostaglandin E2 receptor subtype 4; all P < 5 × 10(-5)). Functional evaluation in regulatory T cells identified that rs4434423A in the PTGER4 gene was associated with differential suppressive function of regulatory T cells. CONCLUSIONS: Further research aimed at replication and additional functional insight into the role played by genetic variation in prostaglandin E2 synthetic and signaling pathways in PGD is warranted.


Subject(s)
Intramolecular Oxidoreductases/genetics , Lung Transplantation , Polymorphism, Single Nucleotide , Primary Graft Dysfunction/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Biomarkers/blood , Computational Biology , Dinoprostone/blood , Female , Genetic Association Studies , Genetic Markers , Genotype , Genotyping Techniques , Humans , Male , Middle Aged , Primary Graft Dysfunction/blood , Primary Graft Dysfunction/immunology , Prospective Studies , Prostaglandin-E Synthases , T-Lymphocytes, Regulatory/metabolism
12.
Liver Transpl ; 20(3): 376-85, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24357103

ABSTRACT

Liver regeneration is a complex process that restores functional tissue after resection or injury, and it is accompanied by transient adenosine triphosphate depletion and metabolic stress in hepatic parenchymal cells. Heat shock protein 70 (Hsp70) functions as a chaperone during periods of cellular stress and induces the expression of several inflammatory cytokines identified as key players during early liver regeneration. We, therefore, hypothesized that Hsp70 is required for the initiation of regeneration. Investigations were carried out in a 70% partial hepatectomy mouse model with mice lacking inducible Hsp70 (Hsp70(-/-)). Liver regeneration was assessed postoperatively with the liver weight/body weight (LW/BW) ratio, and sera and tissues were collected for analysis. In addition, the expression of Hsp-related genes was assessed in a cohort of 23 human living donor liver transplantation donors. In mice, the absence of Hsp70 was associated with a reduced postoperative LW/BW ratio, Ki-67 staining, and tumor necrosis factor α (TNF-α) expression in comparison with wild-type mice. TNF-α expression was also reduced in livers from Hsp70(-/-) mice after induction with lipopolysaccharide (1 mg/kg). Clinically, the transcription of multiple Hsp genes (especially Hsp70 family members) was up-regulated after donor hepatectomy. Together, these results suggest that the early phase of successful liver regeneration requires the presence of Hsp70 to induce TNF-α. Further studies are required to determine whether Hsp70 contributes to liver regeneration as a chaperone by stabilizing specific interactions required for growth signaling or as a paracrine inflammatory signal, as can occur in models of shock.


Subject(s)
HSP70 Heat-Shock Proteins/physiology , Hepatectomy/methods , Liver Regeneration , Liver Transplantation , Animals , Body Weight , Cell Proliferation , Cohort Studies , Endotoxins , Humans , Inflammation , Ki-67 Antigen/metabolism , Lipopolysaccharides/metabolism , Liver Diseases/metabolism , Liver Diseases/surgery , Living Donors , Male , Mice , Mice, Transgenic , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
13.
Blood ; 119(11): 2443-51, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22246031

ABSTRACT

Clinical and experimental studies show that inhibition of histone/protein deacetylases (HDAC) can have important anti-neoplastic effects through cytotoxic and proapoptotic mechanisms. There are also increasing data from nononcologic settings that HDAC inhibitors (HDACi) can exhibit useful anti-inflammatory effects in vitro and in vivo, unrelated to cytotoxicity or apoptosis. These effects can be cell-, tissue-, or context-dependent and can involve modulation of specific inflammatory signaling pathways as well as epigenetic mechanisms. We review recent advances in the understanding of how HDACi alter immune and inflammatory processes, with a particular focus on the effects of HDACi on T-cell biology, including the activation and functions of conventional T cells and the unique T-cell subset, composed of Foxp3(+) T-regulatory cells. Although studies are still needed to tease out details of the various biologic roles of individual HDAC isoforms and their corresponding selective inhibitors, the anti-inflammatory effects of HDACi are already promising and may lead to new therapeutic avenues in transplantation and autoimmune diseases.


Subject(s)
Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/enzymology , Humans
14.
J Immunol ; 189(12): 5659-66, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23136201

ABSTRACT

Adaptive immunity requires signals from both the TCR and the costimulatory molecule CD28. These receptors activate multiple signaling pathways, including the cyclin-dependent kinase (CDK) cascade, and antigenic signals in the absence of costimulation result in a tolerant state that is enforced by the CDK inhibitory protein p27kip1. We find that CDK2, the major target of p27kip1, is highly active in T cells that infiltrate and reject cardiac allografts. We used mice genetically deficient for CDK2 to determine whether CDK2 is required for T cell alloimmunity. Blockade of CD28 costimulation alone was unable to inhibit the rejection of cardiac allografts by wild-type recipients. However, targeting this pathway in CDK2-deficient recipients led to long-term allograft survival. CDK2-deficient CD4(+) T cells proliferated normally in response to stimulation in vitro and in vivo, however, genetic, short hairpin RNA, or small molecule-mediated antagonism of CDK2 resulted in decreased production of IL-2 and IFN-γ. In addition, surviving grafts from CDK2-deficient recipients showed increased infiltration of Foxp3(+) regulatory T cells (Treg), and Treg from CDK2-deficient mice exhibited increased suppressive activity in vitro and in an in vivo model of inflammatory bowel disease. These data suggest that p27kip1 promotes peripheral tolerance through its ability to inhibit CDK2, which otherwise acts to promote conventional T cell differentiation and restrict Treg function.


Subject(s)
Cyclin-Dependent Kinase 2/physiology , Animals , Cells, Cultured , Colitis/enzymology , Colitis/immunology , Colitis/pathology , Cyclin-Dependent Kinase 2/deficiency , Cyclin-Dependent Kinase 2/genetics , Disease Models, Animal , Female , Immune Tolerance/genetics , Lymphocyte Culture Test, Mixed , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
15.
J Immunol ; 189(3): 1330-9, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22732590

ABSTRACT

Adoptive immunotherapy using cultured T cells holds promise for the treatment of cancer and infectious disease. Ligands immobilized on surfaces fabricated from hard materials such as polystyrene plastic are commonly employed for T cell culture. The mechanical properties of a culture surface can influence the adhesion, proliferation, and differentiation of stem cells and fibroblasts. We therefore explored the impact of culture substrate stiffness on the ex vivo activation and expansion of human T cells. We describe a simple system for the stimulation of the TCR/CD3 complex and the CD28 receptor using substrates with variable rigidity manufactured from poly(dimethylsiloxane), a biocompatible silicone elastomer. We show that softer (Young's Modulus [E] < 100 kPa) substrates stimulate an average 4-fold greater IL-2 production and ex vivo proliferation of human CD4(+) and CD8(+) T cells compared with stiffer substrates (E > 2 MPa). Mixed peripheral blood T cells cultured on the stiffer substrates also demonstrate a trend (nonsignificant) toward a greater proportion of CD62L(neg), effector-differentiated CD4(+) and CD8(+) T cells. Naive CD4(+) T cells expanded on softer substrates yield an average 3-fold greater proportion of IFN-γ-producing Th1-like cells. These results reveal that the rigidity of the substrate used to immobilize T cell stimulatory ligands is an important and previously unrecognized parameter influencing T cell activation, proliferation, and Th differentiation. Substrate rigidity should therefore be a consideration in the development of T cell culture systems as well as when interpreting results of T cell activation based upon solid-phase immobilization of TCR/CD3 and CD28 ligands.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Dimethylpolysiloxanes/chemistry , Elastomers/chemistry , Lymphocyte Activation/immunology , Nylons/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Dimethylpolysiloxanes/pharmacology , Elasticity , Elastomers/pharmacology , Humans , Lymphocyte Activation/drug effects , Nylons/pharmacology , Primary Cell Culture/methods , Substrate Specificity/drug effects , Substrate Specificity/immunology
16.
Nat Med ; 13(11): 1299-307, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17922010

ABSTRACT

Histone/protein deacetylases (HDACs) regulate chromatin remodeling and gene expression as well as the functions of more than 50 transcription factors and nonhistone proteins. We found that administration of an HDAC inhibitor (HDACi) in vivo increased Foxp3 gene expression, as well as the production and suppressive function of regulatory T cells (T(reg) cells). Although T(reg) cells express multiple HDACs, HDAC9 proved particularly important in regulating Foxp3-dependent suppression. Optimal T(reg) function required acetylation of several lysines in the forkhead domain of Foxp3, and Foxp3 acetylation enhanced binding of Foxp3 to the Il2 promoter and suppressed endogenous IL-2 production. HDACi therapy in vivo enhanced T(reg)-mediated suppression of homeostatic proliferation, decreased inflammatory bowel disease through T(reg)-dependent effects, and, in conjunction with a short course of low-dose rapamycin, induced permanent, T(reg)-dependent cardiac and islet allograft survival and donor-specific allograft tolerance. Our data show that use of HDACi allows the beneficial pharmacologic enhancement of both the numbers and suppressive function of Foxp3(+) T(reg) cells.


Subject(s)
Cell Differentiation/immunology , Histone Deacetylase Inhibitors , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/enzymology , Animals , Forkhead Transcription Factors/biosynthesis , Histone Deacetylases/biosynthesis , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
17.
Elife ; 122024 Apr 24.
Article in English | MEDLINE | ID: mdl-38655862

ABSTRACT

Ikaros is a transcriptional factor required for conventional T cell development, differentiation, and anergy. While the related factors Helios and Eos have defined roles in regulatory T cells (Treg), a role for Ikaros has not been established. To determine the function of Ikaros in the Treg lineage, we generated mice with Treg-specific deletion of the Ikaros gene (Ikzf1). We find that Ikaros cooperates with Foxp3 to establish a major portion of the Treg epigenome and transcriptome. Ikaros-deficient Treg exhibit Th1-like gene expression with abnormal production of IL-2, IFNg, TNFa, and factors involved in Wnt and Notch signaling. While Ikzf1-Treg-cko mice do not develop spontaneous autoimmunity, Ikaros-deficient Treg are unable to control conventional T cell-mediated immune pathology in response to TCR and inflammatory stimuli in models of IBD and organ transplantation. These studies establish Ikaros as a core factor required in Treg for tolerance and the control of inflammatory immune responses.


Subject(s)
Forkhead Transcription Factors , Gene Expression Regulation , Ikaros Transcription Factor , T-Lymphocytes, Regulatory , Animals , Ikaros Transcription Factor/metabolism , Ikaros Transcription Factor/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Mice , Mice, Knockout
18.
Transplantation ; 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38685198

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury (IRI) causes significant morbidity in liver transplantation among other medical conditions. IRI following liver transplantation contributes to poor outcomes and early graft loss. Histone/protein deacetylases (HDACs) regulate diverse cellular processes, play a role in mediating tissue responses to IRI, and may represent a novel therapeutic target in preventing IRI in liver transplantation. METHODS: Using a previously described standardized model of murine liver warm IRI, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were assessed at 24 and 48 h after reperfusion to determine the effect of different HDAC inhibitors. RESULTS: Broad HDAC inhibition with trichostatin-A (TSA) was protective against hepatocellular damage (P < 0.01 for AST and P < 0.05 for ALT). Although HDAC class I inhibition with MS-275 provided statistically insignificant benefit, tubastatin-A (TubA), an HDAC6 inhibitor with additional activity against HDAC10, provided significant protection against liver IRI (P < 0.01 for AST and P < 0.001 for ALT). Surprisingly genetic deletion of HDAC6 or -10 did not replicate the protective effects of HDAC6 inhibition with TubA, whereas treatment with an HDAC6 BUZ-domain inhibitor, LakZnFD, eliminated the protective effect of TubA treatment in liver ischemia (P < 0.01 for AST and P < 0.01 for ALT). CONCLUSIONS: Our findings suggest TubA, a class IIb HDAC inhibitor, can mitigate hepatic IRI in a manner distinct from previously described class I HDAC inhibition and requires the HDAC6 BUZ-domain activity. Our data corroborate previous findings that HDAC targets for therapeutic intervention of IRI may be tissue-specific, and identify HDAC6 inhibition as a possible target in the treatment of liver IRI.

19.
Front Immunol ; 14: 1301991, 2023.
Article in English | MEDLINE | ID: mdl-38173720

ABSTRACT

Rationale: Sarcoidosis is an inflammatory granulomatous disease of unknown etiology with predominant lung involvement. Organ involvement and disease severity, as well as the nature of immune alterations, vary among patients leading to a range of clinical phenotypes and outcomes. Our objective was to evaluate the association of disease course and immune responses in pulmonary sarcoidosis. Methods: In this prospective cohort study of 30 subjects, most of whom were followed for one year, we evaluated 14 inflammatory markers in plasma, 13 Treg/T cell flow cytometry markers and 8 parameters of FOXP3+ Treg biology, including suppressive function, epigenetic features and stability. Results: We identified a set of 13 immunological parameters that differ in sarcoidosis subjects in comparison with healthy donors. Five of those were inversely correlated with suppressive function of Tregs in sarcoidosis, and six (TNFα, TNFR I and II, sCD25, Ki-67 and number of Tregs) were particularly upregulated or increased in subjects with thoracic lymphadenopathy. Treg suppressive function was significantly lower in patients with thoracic lymphadenopathy, and in patients with higher burdens of pulmonary and systemic symptoms. A combination of five inflammatory markers, Ki-67 expression, Treg function, and lung diffusion capacity evaluated at study entry predicted need for therapy at one year follow-up in 90% of cases. Conclusion: Tregs may suppress ongoing inflammation at local and systemic levels, and TNFα, TNFR I and II, sCD25 and Ki-67 emerge as attractive biomarkers for in vivo sarcoid inflammatory activity.


Subject(s)
Lymphadenopathy , Sarcoidosis , Humans , T-Lymphocytes, Regulatory , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Prospective Studies , Ki-67 Antigen/metabolism , Sarcoidosis/metabolism , Prognosis , Forkhead Transcription Factors/metabolism
20.
Transl Psychiatry ; 13(1): 259, 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37443311

ABSTRACT

The Methyl-CpG-Binding Domain Protein family has been implicated in neurodevelopmental disorders. The Methyl-CpG-binding domain 2 (Mbd2) binds methylated DNA and was shown to play an important role in cancer and immunity. Some evidence linked this protein to neurodevelopment. However, its exact role in neurodevelopment and brain function is mostly unknown. Here we show that Mbd2-deficiency in mice (Mbd2-/-) results in deficits in cognitive, social and emotional functions. Mbd2 binds regulatory DNA regions of neuronal genes in the hippocampus and loss of Mbd2 alters the expression of hundreds of genes with a robust down-regulation of neuronal gene pathways. Further, a genome-wide DNA methylation analysis found an altered DNA methylation pattern in regulatory DNA regions of neuronal genes in Mbd2-/- mice. Differentially expressed genes significantly overlap with gene-expression changes observed in brains of Autism Spectrum Disorder (ASD) individuals. Notably, downregulated genes are significantly enriched for human ortholog ASD risk genes. Observed hippocampal morphological abnormalities were similar to those found in individuals with ASD and ASD rodent models. Hippocampal Mbd2 knockdown partially recapitulates the behavioral phenotypes observed in Mbd2-/- mice. These findings suggest that Mbd2 is a novel epigenetic regulator of genes that are associated with ASD in humans. Mbd2 loss causes behavioral alterations that resemble those found in ASD individuals.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Humans , Animals , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , CpG Islands , Autistic Disorder/genetics , Autism Spectrum Disorder/genetics , DNA Methylation , Cognition , DNA/metabolism , Epigenesis, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL