Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Biol Cell ; 114(7): 185-198, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35389514

ABSTRACT

MAGI-1 is a critical cellular scaffolding protein with over 110 different cellular and microbial protein interactors. Since the discovery of MAGI-1 in 1997, MAGI-1 has been implicated in diverse cellular functions such as polarity, cell-cell communication, neurological processes, kidney function, and a host of diseases including cancer and microbial infection. Additionally, MAGI-1 has undergone nomenclature changes in response to the discovery of an additional PDZ domain, leading to lack of continuity in the literature. We address the nomenclature of MAGI-1 as well as summarize many of the critical functions of the known interactions. Given the importance of many of the interactors, such as human papillomavirus E6, the Coxsackievirus and adenovirus receptor (CAR), and PTEN, the enhancement or disruption of MAGI-based interactions has the potential to affect cellular functions that can potentially be harnessed as a therapeutic strategy for a variety of diseases.


Subject(s)
PDZ Domains , Humans
3.
Clin Transplant ; 34(3): e13822, 2020 03.
Article in English | MEDLINE | ID: mdl-32037573

ABSTRACT

BACKGROUND: Hospital readmission (HR) after surgery is considered a quality metric. METHODS: Data on 2371 first-time adult kidney transplant (KT) recipients were collected to analyze the "early" (≤30 days) and "late" (31-365 days) HR patterns after KT at a single center over a 12-year time span (2002-2013). RESULTS: 30-day, 90-day, and 1-year HR were 31%, 41%, and 53%, respectively. Risk factors for HR included age >50, female sex, black race, BMI >30, transplant LOS >5 days, and pre-transplant time on dialysis >765 days. Indications for early (n = 749) and late (n = 508) HR were similar. Early HR (OR: 3.80, P = .007) and black race (OR: 2.38, P = .009) were associated with higher odds of 1-year graft failure while frequency (1-2, 3-4, 5+) of HR (ORs: 4.68, 8.36, 9.44, P < .001) and age > 50 (OR: 2.11, P = .007) were associated with higher odds of 1-year mortality. Transplant LOS > 5 days increased both odds of 1-year graft failure (OR: 3.51, P = .001) and mortality (OR: 2.05, P = .006). One-year graft and recipient survival were 96.7% and 94.8%, respectively. CONCLUSIONS: Hospital readmission was associated with reduced graft and patient survival; however, despite a relatively high and consistent HR rate after KT, overall 1-year graft and patient survival was high.


Subject(s)
Kidney Transplantation , Adult , Female , Graft Survival , Humans , Patient Readmission , Renal Dialysis , Risk Factors , Transplant Recipients
4.
Transplantation ; 103(9): 1809-1820, 2019 09.
Article in English | MEDLINE | ID: mdl-30830040

ABSTRACT

BACKGROUND: We previously reported the novel activity of alloprimed CD8 T cells that suppress posttransplant alloantibody production. The purpose of the study is to investigate the expression and role of CXCR5 on antibody-suppressor CD8 T-cell function. METHODS: C57BL/6 mice were transplanted with FVB/N hepatocytes. Alloprimed CD8 T cells were retrieved on day 7 from hepatocyte transplant recipients. Unsorted or flow-sorted (CXCR5CXCR3 and CXCR3CXCR5) alloprimed CD8 T-cell subsets were analyzed for in vitro cytotoxicity and capacity to inhibit in vivo alloantibody production following adoptive transfer into C57BL/6 or high alloantibody-producing CD8 knock out (KO) hepatocyte transplant recipients. Alloantibody titer was assessed in CD8 KO mice reconstituted with naive CD8 T cells retrieved from C57BL/6, CXCR5 KO, or CXCR3 KO mice. Antibody suppression by ovalbumin (OVA)-primed monoclonal OVA-specific t-cell receptor transgenic CD8+ T cells (OT-I) CXCR5 or CXCR3 CD8 T-cell subsets was also investigated. RESULTS: Alloprimed CXCR5CXCR3CD8 T cells mediated in vitro cytotoxicity of alloprimed "self" B cells, while CXCR3CXCR5CD8 T cells did not. Only flow-sorted alloprimed CXCR5CXCR3CD8 T cells (not flow-sorted alloprimed CXCR3CXCR5CD8 T cells) suppressed alloantibody production and enhanced graft survival when transferred into transplant recipients. Unlike CD8 T cells from wild-type or CXCR3 KO mice, CD8 T cells from CXCR5 KO mice do not develop alloantibody-suppressor function. Similarly, only flow-sorted CXCR5CXCR3 (and not CXCR3CXCR5) OVA-primed OT-I CD8 T cells mediated in vivo suppression of anti-OVA antibody production. CONCLUSIONS: These data support the conclusion that expression of CXCR5 by antigen-primed CD8 T cells is critical for the function of antibody-suppressor CD8 T cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/prevention & control , Hepatocytes/transplantation , Liver Transplantation , Receptors, CXCR5/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CD8 Antigens/deficiency , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Female , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival , Hepatocytes/immunology , Hepatocytes/metabolism , Liver Transplantation/adverse effects , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Signal Transduction , Time Factors
5.
J Immunol ; 201(12): 3731-3740, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30397035

ABSTRACT

Humoral alloimmunity negatively impacts both short- and long-term cell and solid organ transplant survival. We previously reported that alloantibody-mediated rejection of transplanted hepatocytes is critically dependent on host macrophages. However, the effector mechanism(s) of macrophage-mediated injury to allogeneic liver parenchymal cells is not known. We hypothesized that macrophage-mediated destruction of allogeneic hepatocytes occurs by cell-cell interactions requiring FcγRs. To examine this, alloantibody-dependent hepatocyte rejection in CD8-depleted wild-type (WT) and Fcγ-chain knockout (KO; lacking all functional FcγR) transplant recipients was evaluated. Alloantibody-mediated hepatocellular allograft rejection was abrogated in recipients lacking FcγR compared with WT recipients. We also investigated anti-FcγRI mAb, anti-FcγRIII mAb, and inhibitors of intracellular signaling (to block phagocytosis, cytokines, and reactive oxygen species [ROS]) in an in vitro alloantibody-dependent, macrophage-mediated hepatocytoxicity assay. Results showed that in vitro alloantibody-dependent, macrophage-mediated hepatocytotoxicity was critically dependent on FcγRs and ROS. The adoptive transfer of WT macrophages into CD8-depleted FcγR-deficient recipients was sufficient to induce alloantibody-mediated rejection, whereas adoptive transfer of macrophages from Fcγ-chain KO mice or ROS-deficient (p47 KO) macrophages was not. These results provide the first evidence, to our knowledge, that alloantibody-dependent hepatocellular allograft rejection is mediated by host macrophages through FcγR signaling and ROS cytotoxic effector mechanisms. These results support the investigation of novel immunotherapeutic strategies targeting macrophages, FcγRs, and/or downstream molecules, including ROS, to inhibit humoral immune damage of transplanted hepatocytes and perhaps other cell and solid organ transplants.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Hepatocytes/immunology , Macrophages/immunology , Receptors, IgG/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Cells, Cultured , Cytotoxicity, Immunologic , DNA Helicases/genetics , Humans , Isoantibodies/metabolism , Liver Transplantation , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Receptors, IgG/genetics , Signal Transduction
6.
Transplantation ; 100(9): 1898-906, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27362313

ABSTRACT

BACKGROUND: De novo alloantibodies (donor-specific antibody) contribute to antibody-mediated rejection and poor long-term graft survival. Because the development of donor-specific antibody is associated with early graft loss of cell transplants and reduced long-term survival of solid organ transplants, we hypothesized that conventional immunosuppressives, calcineurin inhibitors (CNi), and mammalian target of rapamycin inhibitors (mTORi), may not be as effective for suppression of humoral alloimmunity as for cell-mediated immunity. METHODS: Wild-type or CD8-depleted mice were transplanted with allogeneic hepatocytes. Recipients were treated with mTORi and/or CNi and serially monitored for alloantibody and graft survival. The direct effect of mTORi and CNi on alloprimed B cell function was investigated in Rag1 mice adoptively transferred with alloprimed IgG1 B cells. The efficacy of mTORi and/or CNi to suppress CD8-mediated cytotoxicity of IgG1 B cells was evaluated in in vitro and in vivo cytotoxicity assays. RESULTS: Mammalian target of rapamycin inhibitors, but not CNi, reduced alloantibody production in transplant recipients, directly suppressed alloantibody production by alloprimed IgG1 B cells and delayed graft rejection in both low and high alloantibody producers. Combination treatment with mTORi and CNi resulted in loss of the inhibitory effect observed for mTORi monotherapy in part due to CNi suppression of CD8 T cells which downregulate alloantibody production (CD8 TAb-supp cells). CONCLUSIONS: Our data support that mTORi is a potent inhibitor of humoral immunity through suppression of alloprimed B cells and preservation of CD8 TAb-supp cells. In contrast, alloantibody is readily detected in CNi-treated recipients because CNi does not suppress alloprimed B cells and interferes with downregulatory CD8 TAb-supp cells.


Subject(s)
B-Lymphocytes/drug effects , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/drug effects , Hepatocytes/transplantation , Immunity, Humoral/drug effects , Immunosuppressive Agents/pharmacology , Isoantibodies/immunology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Calcineurin Inhibitors/pharmacology , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic/drug effects , Down-Regulation , Genotype , Graft Rejection/enzymology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/drug effects , Hepatocytes/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunity, Cellular/drug effects , Isoantibodies/blood , Liver Transplantation/adverse effects , Liver Transplantation/methods , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Phenotype , TOR Serine-Threonine Kinases/metabolism , Time Factors
7.
Transplantation ; 100(9): 1907-15, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27379551

ABSTRACT

BACKGROUND: The liver immune environment is tightly regulated to balance immune activation with immune tolerance. Understanding the dominant immune pathways initiated in the liver is important because the liver is a site for cell transplantation, such as for islet and hepatocyte transplantation. The purpose of this study is to examine the consequences of alloimmune stimulation when allogeneic cells are transplanted to the liver in comparison to a different immune locale, such as the kidney. METHODS: We investigated cellular and humoral immune responses when allogeneic hepatocytes are transplanted directly to the recipient liver by intraportal injection. A heterotopic kidney engraftment site was used for comparison to immune activation in the liver microenvironment. RESULTS: Transplantation of allogeneic hepatocytes delivered directly to the liver, via recipient portal circulation, stimulated long-term, high magnitude CD8 T cell-mediated allocytotoxicity. CD8 T cells initiated significant in vivo allocytotoxicity as well as rapid rejection of hepatocytes transplanted to the liver even in the absence of secondary lymph nodes or CD4 T cells. In contrast, in the absence of recipient peripheral lymphoid tissue and CD4 T cells, CD8-mediated in vivo allocytotoxicity was abrogated, and rejection was delayed when hepatocellular allografts were transplanted to the kidney subcapsular site. CONCLUSIONS: These results highlight the CD8-dominant proinflammatory immune responses unique to the liver microenvironment. Allogeneic cells transplanted directly to the liver do not enjoy immune privilege but rather require immunosuppression to prevent rejection by a robust and persistent CD8-dependent allocytotoxicity primed in the liver.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatocytes/transplantation , Immunity, Cellular , Immunity, Humoral , Liver Transplantation/methods , Liver/surgery , Animals , CD4-Positive T-Lymphocytes/immunology , Cellular Microenvironment , Cytotoxicity, Immunologic , Genotype , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival , Hepatocytes/immunology , Kidney/immunology , Liver/immunology , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Time Factors , Transplantation, Homologous
8.
Sci Rep ; 6: 20646, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26865459

ABSTRACT

Cholesterol from peripheral tissue, carried by HDL, is metabolized in the liver after uptake by the HDL receptor, SR-B1. Hepatocytes have long been considered the only liver cells expressing SR-B1; however, in this study we describe two disparate immunofluorescence (IF) experiments that suggest otherwise. Using high-resolution confocal microscopy employing ultrathin (120 nm) sections of mouse liver, improving z-axis resolution, we identified the liver sinusoidal endothelial cells (LSEC), marked by FcγRIIb, as the cell within the liver expressing abundant SR-B1. In contrast, the hepatocyte, identified with ß-catenin, expressed considerably weaker levels, although optical resolution of SR-B1 was inadequate. Thus, we moved to a different IF strategy, first separating dissociated liver cells by gradient centrifugation into two portions, hepatocytes (parenchymal cells) and LSEC (non-parenchymal cells). Characterizing both portions for the cellular expression of SR-B1 by flow cytometry, we found that LSEC expressed considerable amounts of SR-B1 while in hepatocytes SR-B1 expression was barely perceptible. Assessing mRNA of SR-B1 by real time PCR we found messenger expression in LSEC to be about 5 times higher than in hepatocytes.


Subject(s)
Cholesterol/metabolism , Endothelial Cells/metabolism , Hepatocytes/metabolism , Liver/metabolism , RNA, Messenger/genetics , Scavenger Receptors, Class B/genetics , Animals , Biological Transport , COS Cells , Cell Line , Cell Separation , Chlorocebus aethiops , Endothelial Cells/cytology , Hepatocytes/cytology , Liver/cytology , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microtomy , Organ Specificity , RNA, Messenger/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Scavenger Receptors, Class B/metabolism , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL