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

Country/Region as subject
Affiliation country
Publication year range
1.
Clin Transplant ; 38(2): e15249, 2024 02.
Article in English | MEDLINE | ID: mdl-38369810

ABSTRACT

BACKGROUND: Desensitization is one of the strategies to reduce antibodies and facilitate heart transplantation in highly sensitized patients. We describe our center's desensitization experience with combination of plasma cell (PC) depletion therapy (with proteasome inhibitor or daratumumab) and costimulation blockade (with belatacept). METHODS: We reviewed five highly sensitized patients who underwent desensitization therapy with plasma cell depletion and costimulation blockade. We evaluated the response to therapy by measuring the changes in cPRA, average MFI, and number of positive beads > 5000MFI. RESULTS: Five patients, mean age of 56 (37-66) years with average cPRA of 98% at 5000 MFI underwent desensitization therapy. After desensitization, mean cPRA decreased from 98% to 70% (p = .09), average number of beads > 5000 MFI decreased from 59 to 37 (p = .15), and average MFI of beads > 5000 MFI decreased from 16713 to 13074 (p = .26). CONCLUSION: Combined PC depletion and CoB could be a reasonable strategy for sustained reduction in antibodies in highly sensitized patients being listed for heart transplantation.


Subject(s)
Heart Transplantation , Plasma Cells , Humans , Middle Aged , Abatacept/therapeutic use , Abatacept/pharmacology , Desensitization, Immunologic , Graft Rejection/etiology , Graft Rejection/prevention & control , HLA Antigens , Isoantibodies , Proteasome Inhibitors , Adult , Aged
2.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article in English | MEDLINE | ID: mdl-33785592

ABSTRACT

Natalizumab, a humanized monoclonal antibody (mAb) against α4-integrin, reduces the number of dendritic cells (DC) in cerebral perivascular spaces in multiple sclerosis (MS). Selective deletion of α4-integrin in CD11c+ cells should curtail their migration to the central nervous system (CNS) and ameliorate experimental autoimmune encephalomyelitis (EAE). We generated CD11c.Cre+/-ITGA4fl/fl C57BL/6 mice to selectively delete α4-integrin in CD11c+ cells. Active immunization and adoptive transfer EAE models were employed and compared with WT controls. Multiparameter flow cytometry was utilized to immunophenotype leukocyte subsets. Single-cell RNA sequencing was used to profile individual cells. α4-Integrin expression by CD11c+ cells was significantly reduced in primary and secondary lymphoid organs in CD11c.Cre+/-ITGA4fl/fl mice. In active EAE, a delayed disease onset was observed in CD11c.Cre+/-ITGA4fl/fl mice, during which CD11c+CD88+ cells were sequestered in the blood. Upon clinical EAE onset, CD11c+CD88+ cells appeared in the CNS and expressed CD317+ In adoptive transfer experiments, CD11c.Cre+/-ITGA4fl/fl mice had ameliorated clinical disease phenotype associated with significantly diminished numbers of CNS CD11c+CD88+CD317+ cells. In human cerebrospinal fluid from subjects with neuroinflammation, microglia-like cells display coincident expression of ITGAX (CD11c), C5AR1 (CD88), and BST2 (CD317). In mice, we show that only activated, but not naïve microglia expressed CD11c, CD88, and CD317. Finally, anti-CD317 treatment prior to clinical EAE substantially enhanced recovery in mice.


Subject(s)
Antigens, CD/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/metabolism , Myeloid Cells/metabolism , Animals , Antigen Presentation , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/metabolism , Female , Humans , Male , Mice , Microglia/metabolism
3.
Haematologica ; 108(7): 1840-1850, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-36373249

ABSTRACT

Defects in T-cell immunity to SARS-CoV-2 have been linked to an increased risk of severe COVID-19 (even after vaccination), persistent viral shedding and the emergence of more virulent viral variants. To address this T-cell deficit, we sought to prepare and cryopreserve banks of virus-specific T cells, which would be available as a partially HLA-matched, off-the-shelf product for immediate therapeutic use. By interrogating the peripheral blood of healthy convalescent donors, we identified immunodominant and protective T-cell target antigens, and generated and characterized polyclonal virus-specific T-cell lines with activity against multiple clinically important SARS-CoV-2 variants (including 'delta' and 'omicron'). The feasibility of making and safely utilizing such virus-specific T cells clinically was assessed by administering partially HLA-matched, third-party, cryopreserved SARS-CoV-2-specific T cells (ALVR109) in combination with other antiviral agents to four individuals who were hospitalized with COVID-19. This study establishes the feasibility of preparing and delivering off-the-shelf, SARS-CoV-2-directed, virus-specific T cells to patients with COVID-19 and supports the clinical use of these products outside of the profoundly immune compromised setting (ClinicalTrials.gov number, NCT04401410).


Subject(s)
COVID-19 , Hematopoietic Stem Cell Transplantation , Humans , Lymphocytes , SARS-CoV-2
4.
Clin Transplant ; 37(8): e14963, 2023 08.
Article in English | MEDLINE | ID: mdl-36938669

ABSTRACT

INTRODUCTION: In an effort to maximize living donor kidney utilization, we describe the use of deceased donor vein extension grafts for right-sided living donor kidneys and report our single-center experience using this technique. METHODS: A retrospective review of kidney transplant recipients (KTR) who received a right living donor kidney with deceased donor vein extension graft. Recipient demographics, postoperative graft function, and surgical complications were reviewed. Living donor nephrectomies were performed laparoscopically. Vein grafts were obtained from recent deceased donor procurements. End-to-end anastomosis of the graft to the renal vein was performed prior to implantation. RESULTS: Thirty-eight KTR received a right kidney transplant with deceased donor extension grafts. The median recipient age and BMI were 53.0 years and 29.3 kg/m2 . Total 71% were male. Ninety-five percent of grafts displayed immediate graft function, with two recipients requiring temporary dialysis due to anaphylaxis from induction therapy. Median serum creatinine at two weeks was 1.6 mg/dL and at three months was 1.5 mg/dL. There were no graft thromboses. CONCLUSION: Utilization of deceased donor extension grafts for short right renal veins is a simple technique that expands the donor pool for living donor renal transplantation. Our experience resulted in no technical complications and excellent early graft function.


Subject(s)
Kidney Transplantation , Male , Humans , Female , Kidney Transplantation/methods , Living Donors , Graft Survival , Kidney , Renal Veins/surgery , Retrospective Studies , Treatment Outcome
5.
J Pediatr Gastroenterol Nutr ; 77(3): 354-357, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37347142

ABSTRACT

Non-caseating granulomas may indicate a more aggressive phenotype of Crohn disease (CD). Genetic associations of granulomatous CD (GCD) may help elucidate disease pathogenesis. Whole-exome sequencing was performed on peripheral blood-derived DNA from 17 pediatric patients with GCD and 19 with non-GCD (NGCD), and from an independent validation cohort of 44 GCD and 19 NGCD cases. PLINK (a tool set for whole-genome association and population-based linkage analyses) analysis was used to identify single nucleotide polymorphisms (SNPs) differentiating between groups, and subgroup allele frequencies were also compared to a public genomic database (gnomAD). The Combined Annotation Dependent Depletion scoring tool was used to predict deleteriousness of SNPs. Human leukocyte antigen (HLA) haplotype findings were compared to a control group (n = 8496). PLINK-based analysis between GCD and NGCD groups did not find consistently significant hits. gnomAD control comparisons, however, showed consistent subgroup associations with DGKZ , ESRRA , and GXYLT1 , genes that have been implicated in mammalian granulomatous inflammation. Our findings may guide future research and precision medicine.


Subject(s)
Crohn Disease , Child , Humans , Crohn Disease/complications , Exome Sequencing , Genetic Predisposition to Disease , Granuloma/genetics , Granuloma/pathology , Phenotype , ERRalpha Estrogen-Related Receptor
6.
Am J Pathol ; 191(1): 90-107, 2021 01.
Article in English | MEDLINE | ID: mdl-33157066

ABSTRACT

Coronavirus disease 2019 (COVID-19) convalescent plasma has emerged as a promising therapy and has been granted Emergency Use Authorization by the US Food and Drug Administration for hospitalized COVID-19 patients. We recently reported results from interim analysis of a propensity score-matched study suggesting that early treatment of COVID-19 patients with convalescent plasma containing high-titer anti-spike protein receptor binding domain (RBD) IgG significantly decreases mortality. We herein present results from a 60-day follow-up of a cohort of 351 transfused hospitalized patients. Prospective determination of enzyme-linked immunosorbent assay anti-RBD IgG titer facilitated selection and transfusion of the highest titer units available. Retrospective analysis by the Ortho VITROS IgG assay revealed a median signal/cutoff ratio of 24.0 for transfused units, a value far exceeding the recent US Food and Drug Administration-required cutoff of 12.0 for designation of high-titer convalescent plasma. With respect to altering mortality, our analysis identified an optimal window of 44 hours after hospitalization for transfusing COVID-19 patients with high-titer convalescent plasma. In the aggregate, the analysis confirms and extends our previous preliminary finding that transfusion of COVID-19 patients soon after hospitalization with high-titer anti-spike protein RBD IgG present in convalescent plasma significantly reduces mortality.


Subject(s)
COVID-19/mortality , COVID-19/therapy , Immunoglobulin G/immunology , Spike Glycoprotein, Coronavirus/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Female , Follow-Up Studies , Hospitalization , Humans , Immunization, Passive , Kaplan-Meier Estimate , Linear Models , Male , Middle Aged , Propensity Score , Proportional Hazards Models , Retrospective Studies , Risk , SARS-CoV-2 , Treatment Outcome , COVID-19 Serotherapy
7.
Clin Transplant ; 36(5): e14600, 2022 05.
Article in English | MEDLINE | ID: mdl-35083796

ABSTRACT

Response to two doses of a nucleoside-modified messenger ribonucleic acid (mRNA) vaccine was evaluated in a large solid-organ transplant program. mRNA COVID-19 vaccine was administered to transplant candidates and recipients who met study inclusion criteria. Qualitative anti-SARS-CoV-2 Spike Total Immunoglobulin (Ig) and IgG-specific assays, and a semi-quantitative test for anti-SARS-CoV-2 Spike protein IgG were measured in 241 (17.2%) transplant candidates and 1163 (82.8%) transplant recipients; 55.2% of whom were non-Hispanic White and 44.8% identified as another race. Transplant recipients were a median (IQR) of 3.2 (1.1, 6.8) years from transplantation. Response differed by transplant status: 96.0% versus 43.2% by the anti-SARS-CoV-2 Total Ig (candidates vs. recipients, respectively), 93.5% versus 11.6% by the anti-SARS-CoV-2 IgG assay, and 91.9% versus 30.1% by anti-spike titers after two doses of vaccine. Multivariable analysis revealed candidates had higher likelihood of response versus recipients (odds ratio [OR], 14.6; 95 %CI 2.19, 98.11; P = .02). A slightly lower response was demonstrated in older patients (OR .96; 95 %CI .94, .99; P = .002), patients taking antimetabolites (OR, .21; 95% CI .08, .51; P = .001). Vaccination prior to transplantation should be encouraged.


Subject(s)
COVID-19 , Organ Transplantation , Aged , Antibodies, Viral , BNT162 Vaccine , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Immunity, Humoral , Immunoglobulin G , RNA, Messenger , SARS-CoV-2 , Transplant Recipients
8.
Am J Pathol ; 190(11): 2290-2303, 2020 11.
Article in English | MEDLINE | ID: mdl-32795424

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, has spread globally, and proven treatments are limited. Transfusion of convalescent plasma collected from donors who have recovered from COVID-19 is among many approaches being studied as potentially efficacious therapy. We are conducting a prospective, propensity score-matched study assessing the efficacy of COVID-19 convalescent plasma transfusion versus standard of care as treatment for severe and/or critical COVID-19. We present herein the results of an interim analysis of 316 patients enrolled at Houston Methodist hospitals from March 28 to July 6, 2020. Of the 316 transfused patients, 136 met a 28-day outcome and were matched to 251 non-transfused control COVID-19 patients. Matching criteria included age, sex, body mass index, comorbidities, and baseline ventilation requirement 48 hours from admission, and in a second matching analysis, ventilation status at day 0. Variability in the timing of transfusion relative to admission and titer of antibodies of plasma transfused allowed for analysis in specific matched cohorts. The analysis showed a significant reduction (P = 0.047) in mortality within 28 days, specifically in patients transfused within 72 hours of admission with plasma with an anti-spike protein receptor binding domain titer of ≥1:1350. These data suggest that treatment of COVID-19 with high anti-receptor binding domain IgG titer convalescent plasma is efficacious in early-disease patients.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/mortality , Plasma/immunology , Pneumonia, Viral/mortality , Adult , Aged , Aged, 80 and over , Blood Component Transfusion/methods , COVID-19 , Coronavirus Infections/diagnosis , Coronavirus Infections/therapy , Coronavirus Infections/virology , Female , Humans , Immunization, Passive/mortality , Male , Middle Aged , Pandemics , Plasma/virology , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , Prospective Studies , SARS-CoV-2 , COVID-19 Serotherapy
9.
Am J Pathol ; 190(8): 1680-1690, 2020 08.
Article in English | MEDLINE | ID: mdl-32473109

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, has spread globally, and no proven treatments are available. Convalescent plasma therapy has been used with varying degrees of success to treat severe microbial infections for >100 years. Patients (n = 25) with severe and/or life-threatening COVID-19 disease were enrolled at the Houston Methodist hospitals from March 28, 2020, to April 14, 2020. Patients were transfused with convalescent plasma, obtained from donors with confirmed severe acute respiratory syndrome coronavirus 2 infection who had recovered. The primary study outcome was safety, and the secondary outcome was clinical status at day 14 after transfusion. Clinical improvement was assessed on the basis of a modified World Health Organization six-point ordinal scale and laboratory parameters. Viral genome sequencing was performed on donor and recipient strains. At day 7 after transfusion with convalescent plasma, nine patients had at least a one-point improvement in clinical scale, and seven of those were discharged. By day 14 after transfusion, 19 (76%) patients had at least a one-point improvement in clinical status, and 11 were discharged. No adverse events as a result of plasma transfusion were observed. Whole genome sequencing data did not identify a strain genotype-disease severity correlation. The data indicate that administration of convalescent plasma is a safe treatment option for those with severe COVID-19 disease.


Subject(s)
Coronavirus Infections/therapy , Pneumonia, Viral/therapy , Adult , Aged , Betacoronavirus/genetics , COVID-19 , Female , Humans , Immunization, Passive , Investigational New Drug Application , Male , Middle Aged , Pandemics , SARS-CoV-2 , Texas , Whole Genome Sequencing , Young Adult , COVID-19 Serotherapy
10.
Nat Immunol ; 10(11): 1185-92, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19783989

ABSTRACT

Programmed death 1 (PD-1) is an inhibitory molecule expressed on activated T cells; however, the biological context in which PD-1 controls T cell tolerance remains unclear. Using two-photon laser-scanning microscopy, we show here that unlike naive or activated islet antigen-specific T cells, tolerized islet antigen-specific T cells moved freely and did not swarm around antigen-bearing dendritic cells (DCs) in pancreatic lymph nodes. Inhibition of T cell antigen receptor (TCR)-driven stop signals depended on continued interactions between PD-1 and its ligand, PD-L1, as antibody blockade of PD-1 or PD-L1 resulted in lower T cell motility, enhanced T cell-DC contacts and caused autoimmune diabetes. Blockade of the immunomodulatory receptor CTLA-4 did not alter T cell motility or abrogate tolerance. Thus, PD-1-PD-L1 interactions maintain peripheral tolerance by mechanisms fundamentally distinct from those of CTLA-4.


Subject(s)
Antigens, Surface/immunology , Apoptosis Regulatory Proteins/immunology , B7-1 Antigen/immunology , Immune Tolerance , Membrane Glycoproteins/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/metabolism , B7-1 Antigen/metabolism , B7-H1 Antigen , CTLA-4 Antigen , Cell Movement , Dendritic Cells/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Lymphocyte Activation , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Peptides/metabolism , Programmed Cell Death 1 Receptor
11.
Clin Transplant ; 31(8)2017 08.
Article in English | MEDLINE | ID: mdl-28582797

ABSTRACT

BACKGROUND: De novo donor-specific antibodies (dnDSA) after renal transplant are associated with acute rejection (AR) and graft loss, yet most recipients with dnDSA have stable function and no AR. We assessed whether the persistence of dnDSA increased the risk of a detrimental outcome. METHODS: A single-center review of renal transplant recipients monitored for dnDSA at multiple time points post-transplant. An Isolated dnDSA was defined as one positive dnDSA and no additional positive tests, whereas ≥2 positive dnDSA was defined as persistent dnDSA. RESULTS: Of 708 recipients, 22% developed dnDSA, of whom 64% had persistent dnDSA. At median follow-up of 35 (range 12-74) months, there were fewer episodes of AR in the isolated dnDSA vs the persistent dnDSA group (2% vs 22%; P<.001,) and fewer graft losses with isolated dnDSA vs persistent dnDSA (0% vs 10%; P=.03). Within the persistent dnDSA group, recipients with dnDSA ≥60% of time points, had more AR (32% vs 16%, P=.10) and more graft losses (21% vs 2%; P=.003) than those with dnDSA<60%. CONCLUSIONS: Persistence of dnDSA resulted in more AR and graft failure than a single positive value. Recipients with longer duration of dnDSA persistence had an additional increased risk of AR and graft failure.


Subject(s)
Graft Rejection/immunology , Isoantibodies/immunology , Kidney Transplantation , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Follow-Up Studies , Graft Rejection/diagnosis , Graft Rejection/epidemiology , Graft Survival/immunology , Humans , Kaplan-Meier Estimate , Logistic Models , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Risk Factors
12.
Clin Transplant ; 31(8)2017 08.
Article in English | MEDLINE | ID: mdl-28658512

ABSTRACT

BACKGROUND: The natural history of de novo donor-specific antibodies (dnDSA) after lung transplantation is not well-described. We sought to determine the incidence and risk factors associated with dnDSA and compare outcomes between recipients with transient (or isolated) vs persistent dnDSA after transplantation. METHODS: A single-center review of all lung transplants from 1/2009-7/2013. DSAs were tested eight times in the first year and every 4 months thereafter. Outcomes examined included acute rejection and graft failure. RESULTS: Median follow-up was 18 months (range: 1-61 months), and 24.6% of 333 first-time lung-only transplant recipients developed a dnDSA. Ethnicity, HLA-DQ mismatches, post-transplantation platelet transfusion and Lung Allocation Score >60 were associated with dnDSA (P<.05). Overall graft survival was worse for dnDSA-positive vs negative recipients (P=.025). Of 323 recipients with 1-year follow-up, 72 (22.2%) developed dnDSA, and in 25 (34.7%), the dnDSA was transient and cleared. Recipients with transient dnDSA were less likely to develop acute rejection than those with persistent dnDSA (P=.007). CONCLUSIONS: Early post-lung transplantation, dnDSA occurred in 1/4 of recipients, was associated with peri-transplant risk factors and resulted in decreased survival. Spontaneous clearance of dnDSA, seen in one-third of recipients, was associated with a lower risk of acute rejection.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Isoantibodies/immunology , Lung Transplantation , Adult , Aged , Case-Control Studies , Female , Follow-Up Studies , Graft Rejection/epidemiology , Graft Rejection/therapy , HLA Antigens/immunology , Humans , Kaplan-Meier Estimate , Logistic Models , Male , Middle Aged , Outcome Assessment, Health Care , Retrospective Studies , Risk Factors , Tissue Donors
13.
Cytokine ; 74(1): 5-17, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25458968

ABSTRACT

CD4(+) T helper (Th) cells are critical for proper immune cell homeostasis and host defense, but are also major contributors to pathology of autoimmune and inflammatory diseases. Since the discovery of the Th1/Th2 dichotomy, many additional Th subsets were discovered, each with a unique cytokine profile, functional properties, and presumed role in autoimmune tissue pathology. This includes Th1, Th2, Th17, Th22, Th9, and Treg cells which are characterized by specific cytokine profiles. Cytokines produced by these Th subsets play a critical role in immune cell differentiation, effector subset commitment, and in directing the effector response. Cytokines are often categorized into proinflammatory and anti-inflammatory cytokines and linked to Th subsets expressing them. This article reviews the different Th subsets in terms of cytokine profiles, how these cytokines influence and shape the immune response, and their relative roles in promoting pathology in autoimmune and inflammatory diseases. Furthermore, we will discuss whether Th cell pathogenicity can be defined solely based on their cytokine profiles and whether rigid definition of a Th cell subset by its cytokine profile is helpful.


Subject(s)
Autoimmune Diseases/immunology , Cytokines/immunology , Inflammation/immunology , T-Lymphocyte Subsets/immunology , CD4-Positive T-Lymphocytes/immunology , Humans , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
14.
Hum Immunol ; 84(4): 278-285, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36868898

ABSTRACT

Although rare, infection and vaccination can result in antibodies to human leukocyte antigens (HLA). We analyzed the effect of SARS-CoV-2 infection or vaccination on HLA antibodies in waitlisted renal transplant candidates. Specificities were collected and adjudicated if the calculated panel reactive antibodies (cPRA) changed after exposure. Of 409 patients, 285 (69.7 %) had an initial cPRA of 0 %, and 56 (13.7 %) had an initial cPRA > 80 %. The cPRA changed in 26 patients (6.4 %), 16 (3.9 %) increased, and 10 (2.4 %) decreased. Based on cPRA adjudication, cPRA differences generally resulted from a small number of specificities with subtle fluctuations around the borderline of the participating centers' cutoff for unacceptable antigen listing. All five COVID recovered patients with an increased cPRA were female (p = 0.02). In summary, exposure to this virus or vaccine does not increase HLA antibody specificities and their MFI in approximately 99 % of cases and 97 % of sensitized patients. These results have implications for virtual crossmatching at the time of organ offer after SARS-CoV-2 infection or vaccination, and these events of unclear clinical significance should not influence vaccination programs.


Subject(s)
COVID-19 , Kidney Transplantation , Humans , Female , Male , Tissue Donors , Histocompatibility Testing/methods , Kidney Transplantation/methods , SARS-CoV-2 , Antibodies , HLA Antigens , Vaccination , Isoantibodies
15.
J Exp Med ; 203(12): 2737-47, 2006 Nov 27.
Article in English | MEDLINE | ID: mdl-17116737

ABSTRACT

The past decade has seen a significant increase in the number of potentially tolerogenic therapies for treatment of new-onset diabetes. However, most treatments are antigen nonspecific, and the mechanism for the maintenance of long-term tolerance remains unclear. In this study, we developed an antigen-specific therapy, insulin-coupled antigen-presenting cells, to treat diabetes in nonobese diabetic mice after disease onset. Using this approach, we demonstrate disease remission, inhibition of pathogenic T cell proliferation, decreased cytokine production, and induction of anergy. Moreover, we show that robust long-term tolerance depends on the programmed death 1 (PD-1)-programmed death ligand (PD-L)1 pathway, not the distinct cytotoxic T lymphocyte-associated antigen 4 pathway. Anti-PD-1 and anti-PD-L1, but not anti-PD-L2, reversed tolerance weeks after tolerogenic therapy by promoting antigen-specific T cell proliferation and inflammatory cytokine production directly in infiltrated tissues. PD-1-PD-L1 blockade did not limit T regulatory cell activity, suggesting direct effects on pathogenic T cells. Finally, we describe a critical role for PD-1-PD-L1 in another powerful immunotherapy model using anti-CD3, suggesting that PD-1-PD-L1 interactions form part of a common pathway to selectively maintain tolerance within the target tissues.


Subject(s)
Antigens, Surface/physiology , Apoptosis Regulatory Proteins/physiology , B7-1 Antigen/physiology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/prevention & control , Insulin/physiology , Membrane Glycoproteins/physiology , Peptides/physiology , Signal Transduction/physiology , Animals , B7-H1 Antigen , Cells, Cultured , Female , Immune Tolerance , Mice , Mice, Inbred NOD , Programmed Cell Death 1 Receptor , Remission Induction
16.
Eur J Immunol ; 40(10): 2942-56, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20812237

ABSTRACT

Recent evidence suggests that B- and T-cell interactions may be paramount in relapsing-remitting MS (RRMS) disease pathogenesis. We hypothesized that memory B-cell pools from RRMS patients may specifically harbor a subset of potent neuro-APC that support neuro-Ag reactive T-cell proliferation and cytokine secretion. To test this hypothesis, we compared CD80 and HLA-DR expression, IL-10 and lymphotoxin-α secretion, neuro-Ag binding capacity, and neuro-Ag presentation by memory B cells from RRMS patients to naïve B cells from RRMS patients and to memory and naïve B cells from healthy donors (HD). We identified memory B cells from some RRMS patients that elicited CD4(+) T-cell proliferation and IFN-γ secretion in response to myelin basic protein and myelin oligodendrocyte glycoprotein. Notwithstanding the fact that the phenotypic parameters that promote efficient Ag presentation were observed to be similar between RRMS and HD memory B cells, a corresponding capability to elicit CD4(+) T-cell proliferation in response to myelin basic protein and myelin oligodendrocyte glycoprotein was not observed in HD memory B cells. Our results demonstrate for the first time that the memory B-cell pool in RRMS harbors neuro-Ag specific B cells that can activate T cells.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Myelin Basic Protein/immunology , Myelin-Associated Glycoprotein/immunology , Adult , Cohort Studies , Female , Flow Cytometry , Humans , Immunophenotyping , Interferon-gamma/blood , Interferon-gamma/immunology , Lymphocyte Activation , Lymphotoxin-alpha/immunology , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/immunology , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Young Adult
17.
J Neuroinflammation ; 8: 73, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21702922

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a relevant animal model for the human demyelinating inflammatory disorder of the central nervous system (CNS), multiple sclerosis (MS). Induction of EAE by adoptive transfer allows studying the role of the donor T lymphocyte in disease pathogenesis. It has been challenging to reliably induce adoptive transfer EAE in C57BL/6 (H-2b) mice. The goal of this study was to develop a reproducible and high yield protocol for adoptive transfer EAE in C57BL/6 mice. A step-wise experimental approach permitted us to develop a protocol that resulted in a consistent relatively high disease incidence of ~70% in recipient mice. Donor mice were immunized with myelin oligodendrocyte glycoprotein (MOG)p35-55 in complete Freund's adjuvant (CFA) followed by pertussis toxin (PT). Only lymph node cells (LNC) isolated at day 12 post immunization, and restimulated in vitro for 72 hours with 10 µg/mL of MOGp35-55 and 0.5 ng/mL of interleukin-12 (IL-12) were able to transfer disease. The ability of LNC to transfer disease was associated with the presence of inflammatory infiltrates in the CNS at day 12. Interferon gamma (IFNγ) was produced at comparable levels in cell cultures prepared from mice at both day 6 and day 12 post immunization. By contrast, there was a trend towards a negative association between IL-17 and disease susceptibility in our EAE model. The amount of GM-CSF secreted was significantly increased in the culture supernatants from cells collected at day 12 post immunization versus those collected at day 6 post-immunization. Activated CD4+ T cells present in the day 12 LNC cultures maintained expression of the transcription factor T-bet, which has been shown to regulate the expression of the IL-23 receptor. Also, there was an increased prevalence of MOGp35-55-specific CD4+ T cells in day 12 LNC after in vitro re-stimulation. In summary, encephalitogenic LNC that adoptively transfer EAE in C57BL/6 mice were not characterized by a single biomarker in our study, but by a composite of inflammatory markers. Our data further suggest that GM-CSF expression by CD4+ T cells regulated by IL-23 contributes to their encephalitogenicity in our EAE model.


Subject(s)
Adoptive Transfer/methods , CD4-Positive T-Lymphocytes/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Lymph Nodes/cytology , T-Box Domain Proteins/immunology , Animals , Biomarkers/metabolism , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Glycoproteins/administration & dosage , Glycoproteins/immunology , Humans , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Spinal Cord/immunology , Spinal Cord/pathology
18.
Cell Mol Neurobiol ; 31(6): 867-74, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21625960

ABSTRACT

The pathogenesis of Alzheimer's disease (AD) has been strongly associated with the accumulation of amyloid beta (Aß) peptides in brain, and immunotherapy targeting Aß provides potential for AD prevention. A clinical trial in which AD patients were immunized with Aß42 peptide was stopped when 6% of participants showed meningoencephalitis, apparently due to an inflammatory Th1 immune response. Previously, we and other have shown that Aß42 DNA vaccination via gene gun generates a Th2 cellular immune response, which was shown by analyses of the respective antibody isotype profiles. We also determined that in vitro T cell proliferation in response to Aß42 peptide re-stimulation was absent in DNA Aß42 trimer-immunized mice when compared to Aß42 peptide-immunized mice. To further characterize this observation prospectively and longitudinally, we analyzed the immune response in wild-type mice after vaccination with Aß42 trimer DNA and Aß42 peptide with Quil A adjuvant. Wild-type mice were immunized with short-term (1-3× vaccinations) or long-term (6× vacinations) immunization strategies. Antibody titers and isotype profiles of the Aß42 specific antibodies, as well as cytokine profiles and cell proliferation studies from this longitudinal study were determined. Sufficient antibody titers to effectively reduce Aß42, but an absent T cell proliferative response and no IFNγ or IL-17 secretion after Aß42 DNA trimer immunization minimizes the risk of inflammatory activities of the immune system towards the self antigen Aß42 in brain. Therefore, Aß42 DNA trimer immunization has a high probability to be effective and safe to treat patients with early AD.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , DNA/immunology , Epitopes/immunology , Immunization , Th1 Cells/cytology , Th17 Cells/cytology , Alzheimer Disease/immunology , Animals , Cell Proliferation , Cytokines/metabolism , Enzyme-Linked Immunospot Assay , Fluoresceins , Immunity, Humoral , Mice , Succinimides , Th1 Cells/immunology , Th17 Cells/immunology
19.
Brain ; 133(Pt 2): 375-88, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20145049

ABSTRACT

The primary biological function of the endogenous cellular prion protein has remained unclear. We investigated its biological function in the generation of cellular immune responses using cellular prion protein gene-specific small interfering ribonucleic acid in vivo and in vitro. Our results were confirmed by blocking cellular prion protein with monovalent antibodies and by using cellular prion protein-deficient and -transgenic mice. In vivo prion protein gene-small interfering ribonucleic acid treatment effects were of limited duration, restricted to secondary lymphoid organs and resulted in a 70% reduction of cellular prion protein expression in leukocytes. Disruption of cellular prion protein signalling augmented antigen-specific activation and proliferation, and enhanced T cell receptor signalling, resulting in zeta-chain-associated protein-70 phosphorylation and nuclear factor of activated T cells/activator protein 1 transcriptional activity. In vivo prion protein gene-small interfering ribonucleic acid treatment promoted T cell differentiation towards pro-inflammatory phenotypes and increased survival of antigen-specific T cells. Cellular prion protein silencing with small interfering ribonucleic acid also resulted in the worsening of actively induced and adoptively transferred experimental autoimmune encephalomyelitis. Finally, treatment of myelin basic protein(1-11) T cell receptor transgenic mice with prion protein gene-small interfering ribonucleic acid resulted in spontaneous experimental autoimmune encephalomyelitis. Thus, central nervous system autoimmune disease was modulated at all stages of disease: the generation of the T cell effector response, the elicitation of T effector function and the perpetuation of cellular immune responses. Our findings indicate that cellular prion protein regulates T cell receptor-mediated T cell activation, differentiation and survival. Defects in autoimmunity are restricted to the immune system and not the central nervous system. Our data identify cellular prion protein as a regulator of cellular immunological homoeostasis and suggest cellular prion protein as a novel potential target for therapeutic immunomodulation.


Subject(s)
Demyelinating Autoimmune Diseases, CNS/genetics , Gene Silencing/immunology , Prions/genetics , Receptors, Antigen, T-Cell/physiology , Signal Transduction/immunology , Animals , Demyelinating Autoimmune Diseases, CNS/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prions/immunology , RNA, Small Interfering/genetics
20.
Transplant Proc ; 53(6): 1865-1871, 2021.
Article in English | MEDLINE | ID: mdl-34247857

ABSTRACT

BACKGROUND: Renal allograft survival is negatively affected by the development of de novo posttransplant donor-specific antibodies (dnDSA). We sought to determine whether treatment with intravenous immunoglobulin (IVIG) could remove or reduce the intensity of dnDSA. METHODS: A single-center study of 12 recipients with dnDSA and stable function who received IVIG 1 g/kg monthly for 6 months were compared with a contemporaneous cohort of 24 recipients with dnDSA who did not receive IVIG. RESULTS: The median time to first dnDSA was 6 months (interquartile range [IQR], 1-12), and follow-up was 83 months (IQR, 58-94) posttransplant. Resolution of dnDSA occurred in 27% of IVIG vs 46% of control recipients (P = .48). Fifty-eight percent of recipients in both cohorts demonstrated a reduction in the intensity of the dominant DSA at last follow-up (P =1.0). A reduction in the number of dnDSAs occurred in 58% vs 62% of the IVIG and control cohorts, respectively (P = .81). Post-dnDSA, acute rejection occurred in 8% of the IVIG vs 42% in the control group (P = .06). Forty-two percent of IVIG-treated vs 49% of control recipients had a deterioration in function from first dnDSA until most recent follow-up (P = .81). Actuarial graft survivals were equivalent between groups. CONCLUSIONS: IVIG treatment of dnDSA in recipients with stable graft function had no impact on DSA clearance or MFI reduction, but this outcome may also be owing to sample size. Larger studies or alternate dosing regimens may be required to determine if there is any role for the use of IVIG as a treatment for dnDSA.


Subject(s)
Kidney Transplantation , Follow-Up Studies , Graft Rejection/prevention & control , Graft Survival , HLA Antigens , Humans , Immunoglobulins, Intravenous , Isoantibodies , Kidney Transplantation/adverse effects , Retrospective Studies , Transplant Recipients
SELECTION OF CITATIONS
SEARCH DETAIL