Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Hum Immunol ; 83(5): 467-475, 2022 May.
Article in English | MEDLINE | ID: mdl-35183390

ABSTRACT

Complement dependent cytotoxicity crossmatch (CDC-XM) has been the original standard crossmatch test, whereas, flow cytometry crossmatch (FCXM) is an enhanced and highly sensitive crossmatch assay performed to detect donor specific anti-HLA antibodies (DSA). We analyzed American Society for Histocompatibility and Immunogenetics (ASHI) proficiency testing data (2011-2020) and examined the number of laboratories performing CDC-XM vs. FCXM, the overall efficiency of laboratories in reporting ≥80% consensus CDC-XM vs. FCXM result, and reasons for non-consensus results in the two assays. Of 600 crossmatches in each crossmatch category, the percentage of laboratories reporting T cell CDC-XMs reduced from 40% in 2011 to 13% in 2020, T cell anti-human globulin (AHG) CDC-XM reduced from 56% in 2011 to 21% in 2020, and B cell CDC-XM reduced from 51% in 2011 to 20% in 2020. The percentage of laboratories performing T cell and B cell FCXM remained at approximately 80% throughout. CDC-XM performed on par with FCXM in providing a consensus negative result using negative DSA serum, but under-performed in comparison to FCXM in providing a consensus positive result using positive DSA serum. In addition, only minority of CDC-XMs was reported positive in presence of complement fixing DSA. This study shows that non-consensus CDC-XM was always in presence of HLA IgG DSA and that laboratories may be struggling to interpret the low sensitive CDC-XM results, where highly sensitive solid phase multi-antigen or single antigen assay shows the presence of HLA IgG DSA in serum.


Subject(s)
Kidney Transplantation , Laboratories , Flow Cytometry , Graft Rejection , HLA Antigens , Histocompatibility Testing/methods , Humans , Immunoglobulin G , Isoantibodies
2.
Exp Clin Transplant ; 20(2): 180-189, 2022 02.
Article in English | MEDLINE | ID: mdl-34981713

ABSTRACT

OBJECTIVES: A T-cell-positive and B-cell-negative flow cytometry crossmatch result remains a conundrum since HLA class I antigens are expressed on both T and B cells. We investigated the frequency, donor HLA specificity of the antibodies, and mechanisms for these crossmatch results. MATERIALS AND METHODS: We analyzed 3073 clinical flow cytometry crossmatch tests performed in an American Society of Histocompatibility and Immunogeneticsaccredited histocompatibility laboratory. The sera associated with the T-cell positive and B-cell negative flow cytometry crossmatches were also tested for donor HLA immunoglobulin G antibodies using LABScreen single antigen assays. RESULTS: Among the 3073 test results, 1963 were T-cell negative and B-cell negative, 811 were T-cell negative and B-cell positive, 274 were T-cell positive and B-cell positive, and 25 were T-cell positive and B-cell negative. The LABScreen single antigen assay detected HLA class I immunoglobulin G donor-specific antibodies in 23 of 25 sera associated with a T-cell positive and B-cell negative flow cytometry crossmatch result, and donorspecific antibodies directed at not only HLA-Cw but also at HLA-A or HLA-B were observed. In addition, we identified that the B-cell channel shift threshold used to classify a B-cell flow cytometry crossmatch was a potential contributor to a T-cell-positive and B-cellnegative flow cytometry crossmatch result. CONCLUSIONS: Our analysis of 3073 flow cytometry crossmatches, in addition to demonstrating that HLA antibodies directed at the HLA-A, -B, or -Cw locus were associated with a T-cell-positive and B-cell-negative result, identified mechanisms for the surprising T-cell-positive and B-cell-negative flow cytometry crossmatch result.


Subject(s)
HLA Antigens , T-Lymphocytes , Flow Cytometry/methods , HLA-A Antigens , Histocompatibility Testing/methods , Humans , Immunoglobulin G , Treatment Outcome
4.
Methods Mol Biol ; 1585: 73-82, 2017.
Article in English | MEDLINE | ID: mdl-28477188

ABSTRACT

CD4+ T helper (Th) cell subset generation in vivo requires T cell receptor activation and surface CD28 co-stimulation in the presence of one or more cytokines. Similarly, Th cells can be generated in vitro by activating naïve CD4+CD25- T cells with plate bound-anti-CD3 monoclonal antibody (mAb) (pbCD3) and soluble-anti-CD28 mAb (sCD28) in the presence of polarizing recombinant (r) cytokines and anti-cytokine mAbs. In comparison to in vitro CD4+CD25- T cells, memory CD4+CD25-CD45RO+ T cells have been shown to convert to Th9 cells more efficiently. Here, protocol for in vitro generation of human Th9 cells by activating CD4+CD25-CD45RO+ memory T cells with pbCD3 and sCD28 in the presence of polarizing recombinant interleukin-4 (rIL-4) and transforming growth factor (rTGF-ß) is described.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Antibodies, Monoclonal/metabolism , CD4 Antigens/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-4/metabolism , Interleukin-9/metabolism , Transforming Growth Factor beta/metabolism
5.
Clin Transpl ; 32: 83-91, 2016.
Article in English | MEDLINE | ID: mdl-28564525

ABSTRACT

The relationship between circulating pre-transplant immunoglobulin G (IgG) antibodies to donor human leukocyte antigen (HLA) -C locus determined antigens alone and acute rejection, kidney allograft function, and graft survival is not fully defined. Also, the impact of circulating pre-transplant IgG antibodies to donor HLA-C locus antigens alone on these outcomes has not been compared with the impact of circulating pre-transplant IgG antibodies to donor HLA-A or -B locus antigens. We conducted a retrospective review of records of 1252 kidney allograft recipients transplanted at our center between January 2010 and January 2016 to identify patients with circulating pre-transplant IgG antibodies directed at kidney donor HLA-A, -B, or -C locus determined antigens. Antibodies were detected and reported using the LABScreen Single Antigen Bead assay with microbeads coated with single HLA class I antigens. Pre-transplant and post-transplant data were collected and the graft outcomes of 16 kidney graft recipients with antibodies to HLA-C locus antigens were compared to the outcomes in 56 recipients with antibodies to HLA-A or -B locus determined antigens. The one-year acute rejection rate was 6% in those with donor-specific antibodies (DSA) to HLA-C locus antigens and 20% in those with DSA to HLA-A or -B locus antigens. The graft survival rate was 100% in those with DSA to HLA-C locus antigens and 95% in those with DSA to HLA-A or -B locus antigens. None of the numerical differences were statistically significant (p>0.05). The presence of circulating pre-transplant IgG antibodies directed at kidney donor HLA-C locus antigens alone may not be associated with an increased risk of acute rejection or a decreased graft survival rate. Our observations support the concept that circulating pre-transplant IgG antibodies directed at kidney donor HLA-C locus antigens alone do not negatively impact kidney allograft outcomes and that the mean fluorescence intensities of the antibodies directed at HLA-C locus alone should not be used to list unacceptable HLA-C locus antigens for kidney allocation. A study with a larger cohort is needed to investigate our hypothesis.


Subject(s)
Graft Survival , HLA Antigens , HLA-A Antigens , Isoantibodies , Kidney Transplantation , Allografts , Graft Rejection , Humans , Immunoglobulin G , Retrospective Studies , Tissue Donors
6.
Adv Exp Med Biol ; 809: 103-16, 2014.
Article in English | MEDLINE | ID: mdl-25302368

ABSTRACT

Effective means to identify anti-donor immune activity before the transplant organ is damaged and rejected has been an important goal in transplantation research. Development of sensitive and non-invasive diagnostic methods that probe the immune status of the recipient as well as the resilience of the donor organ should enable personalized application of immunosuppressive drugs. With a non-invasive biomarker for rejection, it should be possible to selectively treat the patients that are rejecting the graft and wean the tolerant patients from immunosuppression. Although A20 is also expressed by activated CD4+ T cells and CD8+ T cells, its expression by mouse tubular cells has been shown to play an important role in protecting allografts from ischemia/reperfusion (I/R) injury and rejection. Using quantitative (real-time) reverse transcriptase polymerase chain reaction (qt-RT-PCR), we showed that expression levels of A20, heme oxygenase (HO)-1, other anti-apoptotic molecules, granzyme-B (GZMB), perforin (PRF1), CD3 and other immune molecules in renal transplant biopsies, urinary cells and peripheral blood cells are predictive of transplantation outcomes. Measuring A20 at mRNA and protein levels has the potentiality to be diagnostic and prognostic of transplantation outcomes and thereby help in timely therapeutic interventions to prolong graft life.


Subject(s)
Biomarkers/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Transplantation , Nuclear Proteins/metabolism , Apoptosis , Humans , Monitoring, Physiologic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Transplantation, Homologous , Tumor Necrosis Factor alpha-Induced Protein 3
8.
PLoS Biol ; 11(7): e1001616, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23935451

ABSTRACT

Cell differentiation is typically directed by external signals that drive opposing regulatory pathways. Studying differentiation under polarizing conditions, with only one input signal provided, is limited in its ability to resolve the logic of interactions between opposing pathways. Dissection of this logic can be facilitated by mapping the system's response to mixtures of input signals, which are expected to occur in vivo, where cells are simultaneously exposed to various signals with potentially opposing effects. Here, we systematically map the response of naïve T cells to mixtures of signals driving differentiation into the Th1 and Th2 lineages. We characterize cell state at the single cell level by measuring levels of the two lineage-specific transcription factors (T-bet and GATA3) and two lineage characteristic cytokines (IFN-γ and IL-4) that are driven by these transcription regulators. We find a continuum of mixed phenotypes in which individual cells co-express the two lineage-specific master regulators at levels that gradually depend on levels of the two input signals. Using mathematical modeling we show that such tunable mixed phenotype arises if autoregulatory positive feedback loops in the gene network regulating this process are gradual and dominant over cross-pathway inhibition. We also find that expression of the lineage-specific cytokines follows two independent stochastic processes that are biased by expression levels of the master regulators. Thus, cytokine expression is highly heterogeneous under mixed conditions, with subpopulations of cells expressing only IFN-γ, only IL-4, both cytokines, or neither. The fraction of cells in each of these subpopulations changes gradually with input conditions, reproducing the continuous internal state at the cell population level. These results suggest a differentiation scheme in which cells reflect uncertainty through a continuously tuneable mixed phenotype combined with a biased stochastic decision rather than a binary phenotype with a deterministic decision.


Subject(s)
T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Separation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , GATA3 Transcription Factor/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , T-Box Domain Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 109(38): 15443-8, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22949661

ABSTRACT

The promise of islet cell transplantation cannot be fully realized in the absence of improvements in engraftment of resilient islets. The marginal mass of islets surviving the serial peritransplant insults may lead to exhaustion and thereby contribute to an unacceptably high rate of intermediate and long-term graft loss. Hence, we have studied the effects of treatment with alpha 1-antitrypsin (AAT) in a syngeneic nonautoimmune islet graft model. A marginal number of syngeneic mouse islets were transplanted into nonautoimmune diabetic hosts and islet function was analyzed in control and AAT treated hosts. In untreated controls, marginal mass islet transplants did not restore euglycemia. Outcomes were dramatically improved by short-term AAT treatment. Transcriptional profiling identified 1,184 differentially expressed transcripts in AAT-treated hosts at 3 d posttransplantation. Systems-biology-based analysis revealed AAT down-regulated regulatory hubs formed by inflammation-related molecules (e.g., TNF-α, NF-κB). The conclusions yielded by the systems-biology analysis were rigorously confirmed by QRT-PCR and immunohistology. These data suggest that short-term AAT treatment of human islet transplant recipients may be worthy of a clinical trial.


Subject(s)
Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , alpha 1-Antitrypsin/metabolism , Animals , Cell Survival , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Immune System , Inflammation , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Treatment Outcome
10.
Eur J Immunol ; 42(11): 3062-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22965858

ABSTRACT

Purinergic signaling and associated ectonucleotidases, such as CD39 and CD73, have been implicated in the pathogenesis of inflammatory bowel disease (IBD). CD39 is known to be a Treg memory cell marker, and here we determine the phenotype and function of CD73(+) CD4(+) T lymphocytes in patients with IBD. We describe elevated levels of CD73(+) CD4(+) T cells in the peripheral blood and intestinal lamina propria of patients with active IBD. The functional phenotype of these CD73(+) CD4(+) T cells was further determined by gene expression, ecto-enzymatic activity, and suppressive assays. Increased numbers of CD73(+) CD4(+) T cells in the periphery and lamina propria were noted during active inflammation, which returned to baseline levels following anti-TNF treatment. Peripheral CD73(+) CD4(+) T cells predominantly expressed CD45RO, and were enriched with IL-17A(+) cells. The CD73(+) CD4(+) cell population expressed higher levels of RORC, IL-17A, and TNF, and lower levels of FOXP3 and/or CD25, than CD73(-) CD4(+) T cells. Expression of CD73 by peripheral CD4(+) T cells was increased by TNF, and decreased by an anti-TNF monoclonal antibody (infliximab). In vitro, these peripheral CD73(+) CD4(+) T cells did not suppress proliferation of CD25(-) effector cells, and expressed higher levels of pro-inflammatory markers. We conclude that the CD73(+) CD4(+) T-cell population in patients with active IBD are enriched with cells with a T-helper type 17 phenotype, and could be used to monitor disease activity during treatment.


Subject(s)
5'-Nucleotidase/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Inflammatory Bowel Diseases/immunology , Mucous Membrane/immunology , Th17 Cells/immunology , 5'-Nucleotidase/blood , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Flow Cytometry , GPI-Linked Proteins/blood , GPI-Linked Proteins/immunology , Gastrointestinal Agents/pharmacology , Gastrointestinal Agents/therapeutic use , Humans , Immunophenotyping , Inflammatory Bowel Diseases/blood , Infliximab , Leukocytes, Mononuclear/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
11.
PLoS One ; 7(5): e33254, 2012.
Article in English | MEDLINE | ID: mdl-22606220

ABSTRACT

BACKGROUND: Previously, we have demonstrated that short-term treatment of new onset diabetic Non-obese diabetic (NOD) mice, mice that are afflicted with both type 1 (T1D) and type 2 (T2D) diabetes with either Power Mix (PM) regimen or alpha1 antitrypsin (AAT) permanently restores euglycemia, immune tolerance to self-islets and normal insulin signaling. METHODOLOGY AND PRINCIPAL FINDINGS: To search for relevant therapeutic targets, we have applied genome wide transcriptional profiling and systems biology oriented bioinformatics analysis to examine the impact of the PM and AAT regimens upon pancreatic lymph node (PLN) and fat, a crucial tissue for insulin dependent glucose disposal, in new onset diabetic non-obese diabetic (NOD) mice. Systems biology analysis identified tumor necrosis factor alpha (TNF-α) as the top focus gene hub, as determined by the highest degree of connectivity, in both tissues. In PLNs and fat, TNF-α interacted with 53% and 32% of genes, respectively, associated with reversal of diabetes by previous treatments and was thereby selected as a therapeutic target. Short-term anti-TNF-α treatment ablated a T cell-rich islet-invasive and beta cell-destructive process, thereby enhancing beta cell viability. Indeed anti-TNF-α treatment induces immune tolerance selective to syngeneic beta cells. In addition to these curative effects on T1D anti-TNF-α treatment restored in vivo insulin signaling resulting in restoration of insulin sensitivity. CONCLUSIONS: In short, our molecular analysis suggested that PM and AAT both may act in part by quenching a detrimental TNF-α dependent effect in both fat and PLNs. Indeed, short-term anti-TNF-α mAb treatment restored enduring euglycemia, self-tolerance, and normal insulin signaling.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 2/etiology , Tumor Necrosis Factor-alpha/physiology , Adipose Tissue/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/therapy , Female , Gene Regulatory Networks , Genome-Wide Association Study , Immune Tolerance , Insulin/blood , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Islets of Langerhans Transplantation , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Systems Biology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , alpha 1-Antitrypsin/pharmacology
12.
J Cell Physiol ; 227(4): 1382-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21618526

ABSTRACT

Inflammation induces the NF-κB dependent protein A20 in human renal proximal tubular epithelial cells (RPTEC), which secondarily contains inflammation by shutting down NF-κB activation. We surmised that inducing A20 without engaging the pro-inflammatory arm of NF-κB could improve outcomes in kidney disease. We showed that hepatocyte growth factor (HGF) increases A20 mRNA and protein levels in RPTEC without causing inflammation. Upregulation of A20 by HGF was NF-κB/RelA dependent as it was abolished by overexpressing IκBα or silencing p65/RelA. Unlike TNFα, HGF caused minimal IκBα and p65/RelA phosphorylation, with moderate IκBα degradation. Upstream, HGF led to robust and sustained AKT activation, which was required for p65 phosphorylation and A20 upregulation. While HGF treatment of RPTEC significantly increased A20 mRNA, it failed to induce NF-κB dependent, pro-inflammatory MCP-1, VCAM-1, and ICAM-1 mRNA. This indicates that HGF preferentially upregulates protective (A20) over pro-inflammatory NF-κB dependent genes. Upregulation of A20 supported the anti-inflammatory effects of HGF in RPTEC. HGF pretreatment significantly attenuated TNFα-mediated increase of ICAM-1, a finding partially reversed by silencing A20. In conclusion, this is the first demonstration that HGF activates an AKT-p65/RelA pathway to preferentially induce A20 but not inflammatory molecules. This could be highly desirable in acute and chronic renal injury where A20-based anti-inflammatory therapies are beneficial.


Subject(s)
Hepatocyte Growth Factor/pharmacology , Inflammation/prevention & control , Intracellular Signaling Peptides and Proteins/genetics , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , NF-kappa B/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Cells, Cultured , DNA-Binding Proteins , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Kidney Tubules, Proximal/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor alpha-Induced Protein 3 , Up-Regulation/drug effects
13.
Clin Transpl ; : 229-35, 2012.
Article in English | MEDLINE | ID: mdl-23721027

ABSTRACT

The predominant anti-HLA IgG subclass during antibody mediated rejection episodes is IgG1. Class I DSA IgG subclasses respond to single cycle of bortezomib-based therapy more frequently as compared to Class II DSA IgG subclasses. High titer Class II DSA IgG subclasses are less likely to respond to single cycle bortezomib-based therapy. Future studies are needed to determine if the response to bortezomib-based therapy is dependent on the type of DSA and/or the strength of the DSA at the time of antibody mediated rejection episode.


Subject(s)
Boronic Acids/therapeutic use , Graft Rejection/drug therapy , Graft Rejection/immunology , HLA Antigens/immunology , Immunoglobulin G/immunology , Organ Transplantation , Pyrazines/therapeutic use , Adult , Antineoplastic Agents/therapeutic use , Bortezomib , Female , Humans , Immunoglobulin G/blood , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Plasmapheresis , Steroids/therapeutic use , Tissue Donors
14.
Nature ; 474(7350): 216-9, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21654805

ABSTRACT

Stem cells reside in a specialized regulatory microenvironment or niche, where they receive appropriate support for maintaining self-renewal and multi-lineage differentiation capacity. The niche may also protect stem cells from environmental insults including cytotoxic chemotherapy and perhaps pathogenic immunity. The testis, hair follicle and placenta are all sites of residence for stem cells and are immune-suppressive environments, called immune-privileged sites, where multiple mechanisms cooperate to prevent immune attack, even enabling prolonged survival of foreign allografts without immunosuppression. We sought to determine if somatic stem-cell niches more broadly are immune-privileged sites by examining the haematopoietic stem/progenitor cell (HSPC) niche in the bone marrow, a site where immune reactivity exists. We observed persistence of HSPCs from allogeneic donor mice (allo-HSPCs) in non-irradiated recipient mice for 30 days without immunosuppression with the same survival frequency compared to syngeneic HSPCs. These HSPCs were lost after the depletion of FoxP3 regulatory T (T(reg)) cells. High-resolution in vivo imaging over time demonstrated marked co-localization of HSPCs with T(reg) cells that accumulated on the endosteal surface in the calvarial and trabecular bone marrow. T(reg) cells seem to participate in creating a localized zone where HSPCs reside and where T(reg) cells are necessary for allo-HSPC persistence. In addition to processes supporting stem-cell function, the niche will provide a relative sanctuary from immune attack.


Subject(s)
Graft Survival/immunology , Hematopoietic Stem Cells/immunology , Imaging, Three-Dimensional , Stem Cell Niche/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Survival/immunology , Cells, Cultured , Forkhead Transcription Factors/metabolism , Hematopoietic Stem Cells/cytology , Humans , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Stem Cell Niche/cytology , T-Lymphocytes, Regulatory/metabolism , Time Factors , Transplantation, Homologous/immunology
15.
PLoS One ; 6(4): e19144, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559452

ABSTRACT

PURPOSE: Resistance to antiangiogenic therapy is an important clinical problem. We examined whether resistance occurs at least in part via reversible, physiologic changes in the tumor, or results solely from stable genetic changes in resistant tumor cells. EXPERIMENTAL DESIGN: Mice bearing two human RCC xenografts were treated with sorafenib until they acquired resistance. Resistant 786-O cells were harvested and reimplanted into naïve mice. Mice bearing resistant A498 cells were subjected to a 1 week treatment break. Sorafenib was then again administered to both sets of mice. Tumor growth patterns, gene expression, viability, blood vessel density, and perfusion were serially assessed in treated vs control mice. RESULTS: Despite prior resistance, reimplanted 786-O tumors maintained their ability to stabilize on sorafenib in sequential reimplantation steps. A transcriptome profile of the tumors revealed that the gene expression profile of tumors upon reimplantation reapproximated that of the untreated tumors and was distinct from tumors exhibiting resistance to sorafenib. In A498 tumors, revascularization was noted with resistance and cessation of sorafenib therapy and tumor perfusion was reduced and tumor cell necrosis enhanced with re-exposure to sorafenib. CONCLUSIONS: In two RCC cell lines, resistance to sorafenib appears to be reversible. These results support the hypothesis that resistance to VEGF pathway therapy is not solely the result of a permanent genetic change in the tumor or selection of resistant clones, but rather is due to a great extent to reversible changes that likely occur in the tumor and/or its microenvironment.


Subject(s)
Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Carcinoma, Renal Cell/drug therapy , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/drug therapy , Pyridines/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Niacinamide/analogs & derivatives , Phenylurea Compounds , Sorafenib
16.
Cell Transplant ; 20(11-12): 1759-69, 2011.
Article in English | MEDLINE | ID: mdl-21439133

ABSTRACT

It has been reported that rapamycin (RPM) can induce de novo conversion of the conventional CD4(+)Foxp3(-) T cells into CD4(+)Foxp3(+) regulatory T cells (iTregs) in transplantation setting. It is not clear whether RPM can similarly generate suppressor CD8(+) T cells to facilitate graft acceptance. In this study, we investigated the ability of short-term RPM treatment in promoting long-term acceptance (LTA) of MHC-mismatched skin allografts by generating a CD8(+) suppressor T-cell population. We found that CD4 knockout (KO) mice (in C57BL/6 background, H-2(b)) can promptly reject DBA/2 (H-2(d)) skin allografts with mean survival time (MST) being 13 days (p < 0.01). However, a short course RPM treatment in these animals induced LTA with graft MST longer than 100 days. Adoptive transfer of CD8(+) T cells from LTA group into recombination-activating gene 1 (Rag-1)-deficient mice provided donor-specific protection of DBA/2 skin grafts against cotransferred conventional CD8(+) T cells. Functionally active immunoregulatory CD8(+) T cells also resided in donor skin allografts. Eighteen percent of CD8(+) suppressor T cells expressed CD28 as measured by flow cytometry, and produced reduced levels of IFN-γ, IL-2, and IL-10 in comparison to CD8(+) effector T cells as measured by ELISA. It is unlikely that CD8(+) suppressor T cells mediated graft protection via IL-10, as IL-10/Fc fusion protein impaired RPM-induced LTA in CD4 KO mice. Our data supported the notion that RPM-induced suppressor CD8(+) T cells home to the allograft and exert donor-specific graft protection.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Graft Rejection/prevention & control , Immunosuppressive Agents/pharmacology , Sirolimus/pharmacology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Graft Rejection/mortality , Graft Survival/drug effects , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/pathology , Skin Transplantation , Transplantation, Homologous
17.
PLoS One ; 5(12): e14240, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-21151899

ABSTRACT

BACKGROUND: Accelerated atherosclerosis is the leading cause of morbidity and mortality in diabetic patients. Hyperglycemia is a recognized independent risk factor for heightened atherogenesis in diabetes mellitus (DM). However, our understanding of the mechanisms underlying glucose damage to the vasculature remains incomplete. METHODOLOGY/PRINCIPAL FINDINGS: High glucose and hyperglycemia reduced upregulation of the NF-κB inhibitory and atheroprotective protein A20 in human coronary endothelial (EC) and smooth muscle cell (SMC) cultures challenged with Tumor Necrosis Factor alpha (TNF), aortae of diabetic mice following Lipopolysaccharide (LPS) injection used as an inflammatory insult and in failed vein-grafts of diabetic patients. Decreased vascular expression of A20 did not relate to defective transcription, as A20 mRNA levels were similar or even higher in EC/SMC cultured in high glucose, in vessels of diabetic C57BL/6 and FBV/N mice, and in failed vein grafts of diabetic patients, when compared to controls. Rather, decreased A20 expression correlated with post-translational O-Glucosamine-N-Acetylation (O-GlcNAcylation) and ubiquitination of A20, targeting it for proteasomal degradation. Restoring A20 levels by inhibiting O-GlcNAcylation, blocking proteasome activity, or overexpressing A20, blocked upregulation of the receptor for advanced glycation end-products (RAGE) and phosphorylation of PKCßII, two prime atherogenic signals triggered by high glucose in EC/SMC. A20 gene transfer to the aortic arch of diabetic ApoE null mice that develop accelerated atherosclerosis, attenuated vascular expression of RAGE and phospho-PKCßII, significantly reducing atherosclerosis. CONCLUSIONS: High glucose/hyperglycemia regulate vascular A20 expression via O-GlcNAcylation-dependent ubiquitination and proteasomal degradation. This could be key to the pathogenesis of accelerated atherosclerosis in diabetes.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/metabolism , Cysteine Endopeptidases/genetics , Diabetes Mellitus, Experimental/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Ubiquitin/chemistry , Animals , Cysteine Endopeptidases/metabolism , Glycosylation , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , NF-kappa B/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3
18.
Hum Immunol ; 71(5): 451-5, 2010 May.
Article in English | MEDLINE | ID: mdl-20156509

ABSTRACT

We have previously demonstrated that biomarkers of inflammation and immune activity detected within intraoperative renal transplant allograft biopsies are linked to adverse short-term post-transplantation clinical outcomes. Now we provide a post hoc analysis of our earlier data in the light of longer clinical follow-up. A total of 75 consecutively performed renal allografts were analyzed for gene expression of proinflammatory molecules, inflammation-induced adhesion molecules, and antiapoptotic genes expressed 15 minutes after vascular reperfusion to determine whether this analysis can aid in predicting long-term quality of renal function, proteinuria, graft loss, and death-censored graft. We have built predictive models for proteinuria (area under the curve = 0.859, p = 0.0001) and graft loss (area under the curve = 0.724, p = 0.027) 2 years post-transplantation using clinical variables in combination with intragraft gene expression data of tumor necrosis factor-alpha, interleukin-6, CD40, CD3, and tumor necrosis factor-alpha, Bcl-2, and interferon-gamma, respectively. This post hoc analysis demonstrates that hypothesis-driven, targeted polymerase chain reaction profiling of gene expression in the donor kidney at the time of engraftment can predict 2-year post-transplantation clinical outcomes.


Subject(s)
Biomarkers/analysis , Gene Expression Profiling , Graft Survival/genetics , Kidney Transplantation/immunology , Tissue Donors , Area Under Curve , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/immunology , Humans , Polymerase Chain Reaction , ROC Curve , Treatment Outcome
19.
PLoS One ; 5(1): e8706, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-20090929

ABSTRACT

BACKGROUND: IL-9 is a growth factor for T- and mast-cells that is secreted by human Th2 cells. We recently reported that IL-4+TGF-beta directs mouse CD4(+)CD25(-)CD62L(+) T cells to commit to inflammatory IL-9 producing CD4(+) T cells. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that human inducible regulatory T cells (iTregs) also express IL-9. IL-4+TGF-beta induced higher levels of IL-9 expression in plate bound-anti-CD3 mAb (pbCD3)/soluble-anti-CD28 mAb (sCD28) activated human resting memory CD4(+)CD25(-)CD45RO(+) T cells as compared to naïve CD4(+)CD25(-)CD45RA(+) T cells. In addition, as compared to pbCD3/sCD28 plus TGF-beta stimulation, IL-4+TGF-beta stimulated memory CD4(+)CD25(-)CD45RO(+) T cells expressed reduced FOXP3 protein. As analyzed by pre-amplification boosted single-cell real-time PCR, human CD4(+)IL-9(+) T cells expressed GATA3 and RORC, but not IL-10, IL-13, IFNgamma or IL-17A/F. Attempts to optimize IL-9 production by pbCD3/sCD28 and IL-4+TGF-beta stimulated resting memory CD4(+) T cells demonstrated that the addition of IL-1beta, IL-12, and IL-21 further enhance IL-9 production. CONCLUSIONS/SIGNIFICANCE: Taken together these data show both the differences and similarities between mouse and human CD4(+)IL9(+) T cells and reaffirm the powerful influence of inflammatory cytokines to shape the response of activated CD4(+) T cells to antigen.


Subject(s)
CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Interleukin-9/immunology , Cytokines/administration & dosage , Enzyme-Linked Immunosorbent Assay , Humans , Polymerase Chain Reaction
20.
Cell Cycle ; 8(9): 1444-50, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19342884

ABSTRACT

Within the immune system there is an exquisite ability to discriminate between "self" and "non-self" that is orchestrated by T lymphocytes. Discriminatory pathways guide differentiation of these lymphocytes into either regulatory (Treg) or effector (Teff) T cells, influenced by cues from the naïve T cell's immediate micro-environment as it responds to cognate antigen. Reciprocal pathways may lead to commitment of naïve T cells into either the protective tolerance-promoting Treg, or to the pro-inflammatory Th17 effector phenotype. Primary activation of CD4(+) lymphocytes stimulates their release of leukemia inhibitory factor (LIF), and Treg continue to release LIF in response to antigen, implying a role for LIF in tolerance. In contrast, interleukin- 6 (IL-6), although very closely related to LIF, promotes maturation of Th17 cells. Here we show that LIF and IL-6 behave as polar opposites in promoting commitment to the Treg and Th17 lineages. Unlike IL6, LIF supported expression of Foxp3, the Treg lineage transcription factor, and LIF opposed IL6 by suppressing IL-6-induced IL-17A protein release. In striking contrast, we found that IL6 effectively inhibited LIF signalling, repressing transcription of the LIF receptor gp190, and strongly inducing axotrophin/MARCH-7, a novel E3 ubitquitin ligase that we discovered to be active in degradation of gp190 protein. In vivo, anti-LIF treatment reduced donor-specific Treg in recipients of foreign spleen cells. Conversely, a single dose of biodegradable LIF nanoparticles, targeted to CD4, successfully manipulated the LIF/IL6 axis towards development of donor-specific Foxp3(+) Treg. The implications for therapy are profound, harnessing endogenous immune regulation by paracrine delivery of LIF to CD4(+) cells in vivo.


Subject(s)
Cell Lineage/drug effects , Interleukin-6/pharmacology , Leukemia Inhibitory Factor/pharmacology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , Animals , Forkhead Transcription Factors/metabolism , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Mice , Models, Immunological , Phenotype , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...