Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Biochem Biophys Res Commun ; 725: 150215, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38870845

ABSTRACT

Cardiac ischemia results in anaerobic metabolism and lactic acid accumulation and with time, intracellular and extracellular acidosis. Ischemia and subsequent reperfusion injury (IRI) lead to various forms of programmed cell death. Necroptosis is a major form of programmed necrosis that worsens cardiac function directly and also promotes inflammation by the release of cellular contents. Potential effects of increasing acidosis on programmed cell death and their specific components have not been well studied. While apoptosis is caspase-dependent, in contrast, necroptosis is mediated by the receptor-interacting protein kinases 1 and 3 (RIPK1/3). In our study, we observed that at physiological pH = 7.4, caspase-8 inhibition did not prevent TNFα-induced cell death in mouse cardiac vascular endothelial cells (MVECs) but promoted necroptotic cell death. As expected, necroptosis was blocked by RIPK1 inhibition. However, at pH = 6.5, TNFα induced an apoptosis-like pattern which was inhibited by caspase-8 inhibition. Interestingly phosphorylation of necroptotic molecules RIPK1, RIPK3, and mixed lineage kinase domain-like protein (MLKL) was enhanced in an acidic pH environment. However, RIPK3 and MLKL phosphorylation was self-limited which may have limited their participation in necroptosis. In addition, an acidic pH promoted apoptosis-inducing factor (AIF) cleavage and nuclear translocation. AIF RNA silencing inhibited cell death, supporting the role of AIF in this cell death. In summary, our study demonstrated that the pH of the micro-environment during inflammation can bias cell death pathways by altering the function of necroptosis-related molecules and promoting AIF-mediated cell death. Further insights into the mechanisms by which an acidic cellular micro-environment influences these and perhaps other forms of regulated cell death, may lead to therapeutic strategies to attenuate IRI.


Subject(s)
Apoptosis , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Tumor Necrosis Factor-alpha , Animals , Hydrogen-Ion Concentration , Apoptosis/drug effects , Necroptosis/drug effects , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/metabolism , Caspase 8/metabolism , Protein Kinases/metabolism , Protein Kinases/genetics , Cells, Cultured , Phosphorylation , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology
2.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38674016

ABSTRACT

Organ transplantation is associated with various forms of programmed cell death which can accelerate transplant injury and rejection. Targeting cell death in donor organs may represent a novel strategy for preventing allograft injury. We have previously demonstrated that necroptosis plays a key role in promoting transplant injury. Recently, we have found that mitochondria function is linked to necroptosis. However, it remains unknown how necroptosis signaling pathways regulate mitochondrial function during necroptosis. In this study, we investigated the receptor-interacting protein kinase 3 (RIPK3) mediated mitochondrial dysfunction and necroptosis. We demonstrate that the calmodulin-dependent protein kinase (CaMK) family members CaMK1, 2, and 4 form a complex with RIPK3 in mouse cardiac endothelial cells, to promote trans-phosphorylation during necroptosis. CaMK1 and 4 directly activated the dynamin-related protein-1 (Drp1), while CaMK2 indirectly activated Drp1 via the phosphoglycerate mutase 5 (PGAM5). The inhibition of CaMKs restored mitochondrial function and effectively prevented endothelial cell death. CaMKs inhibition inhibited activation of CaMKs and Drp1, and cell death and heart tissue injury (n = 6/group, p < 0.01) in a murine model of cardiac transplantation. Importantly, the inhibition of CaMKs greatly prolonged heart graft survival (n = 8/group, p < 0.01). In conclusion, CaMK family members orchestrate cell death in two different pathways and may be potential therapeutic targets in preventing cell death and transplant injury.


Subject(s)
Dynamins , Graft Rejection , Heart Transplantation , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Mice , Graft Rejection/metabolism , Graft Rejection/pathology , Heart Transplantation/adverse effects , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Dynamins/metabolism , Dynamins/genetics , Mitochondria/metabolism , Endothelial Cells/metabolism , Male , Mice, Inbred C57BL , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/genetics , Phosphorylation , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Signal Transduction
3.
Article in English | MEDLINE | ID: mdl-38421272

ABSTRACT

The immune system is regulated by a complex set of genetic, molecular, and cellular interactions. Rapid advances in the study of immunity and its network of interactions have been boosted by a spectrum of "omics" technologies that have generated huge amounts of data that have reached the status of big data (BD). With recent developments in artificial intelligence (AI), theoretical and clinical breakthroughs could emerge. Analyses of large data sets with AI tools will allow the formulation of new testable hypotheses open new research avenues and provide innovative strategies for regulating immunity and treating immunological diseases. This includes diagnosis and identification of rare diseases, prevention and treatment of autoimmune diseases, allergic disorders, infectious diseases, metabolomic disorders, cancer, and organ transplantation. However, ethical and regulatory challenges remain as to how these studies will be used to advance our understanding of basic immunology and how immunity might be regulated in health and disease. This will be particularly important for entities in which the complexity of interactions occurring at the same time and multiple cellular pathways have eluded conventional approaches to understanding and treatment. The analyses of BD by AI are likely to be complicated as both positive and negative outcomes of regulating immunity may have important ethical ramifications that need to be considered. We suggest there is an immediate need to develop guidelines as to how the analyses of immunological BD by AI tools should guide immune-based interventions to treat various diseases, prevent infections, and maintain health within an ethical framework.


Subject(s)
Autoimmune Diseases , Hypersensitivity , Humans , Artificial Intelligence , Big Data , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Cell Communication
4.
Transpl Infect Dis ; 25(4): e14081, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37247212

ABSTRACT

BACKGROUND: Delayed graft function (DGF) increases the renal allograft failure risk. Late-onset Cytomegalovirus (CMV) infection's effect on the association between DGF and allograft failure has not been determined. METHODS: In this retrospective cohort, we included all renal allograft recipients at London Health Sciences Centre from January 1, 2014 to December 30, 2017, and continued clinical follow-up until February 28, 2020. We determined whether late-onset CMV infection affects the association between DGF and allograft failure in stratified and Cox proportional hazard analyses. RESULTS: Of 384 patients (median age [interquartile range]: 55 [43.3-63]; 38.7% female), 57 recipients (14.8%) were diagnosed with DGF. Patients with DGF were at a greater risk of CMV infection than patients without DGF (22.8% vs. 11.3%, p = .017). Late-onset CMV infection (odds ratio [OR]: 4.7, 95% CI: 2.07-10.68) and rejection (OR: 9.59, 95% CI: 4.15-22.16) significantly increased the risk of allograft failure in recipients with DGF. Patients with DGF had a significantly greater risk of graft failure than those without DGF (17.5% vs. 6.1%, p = .007). In the adjusted Cox hazard model, CMV infection significantly increased the risk of allograft failure (aHR: 3.19, 95% CI: 1.49-6.84). CONCLUSION: Late-onset CMV infection considerably increased the risk of graft failure in patients with DGF. A hybrid preventive model including prophylaxis followed by CMV-specific cell-mediated immunity monitoring may decrease the risk of allograft failure in recipients with DGF.


Subject(s)
Cytomegalovirus Infections , Kidney Transplantation , Renal Insufficiency , Humans , Female , Male , Kidney Transplantation/adverse effects , Retrospective Studies , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/epidemiology , Kidney , Cytomegalovirus , Disease Progression , Allografts
5.
Transplantation ; 107(7): 1630-1641, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36949034

ABSTRACT

BACKGROUND: Urine CXCL10 (C-X-C motif chemokine ligand 10, interferon gamma-induced protein 10 [IP10]) outperforms standard-of-care monitoring for detecting subclinical and early clinical T-cell-mediated rejection (TCMR) and may advance TCMR therapy development through biomarker-enriched trials. The goal was to perform an international multicenter validation of a CXCL10 bead-based immunoassay (Luminex) for transplant surveillance and compare with an electrochemiluminescence-based (Meso Scale Discovery [MSD]) assay used in transplant trials. METHODS: Four laboratories participated in the Luminex assay development and evaluation. Urine CXCL10 was measured by Luminex and MSD in 2 independent adult kidney transplant trial cohorts (Basel and TMCT04). In an independent test and validation set, a linear mixed-effects model to predict (log 10 -transformed) MSD CXCL10 from Luminex CXCL10 was developed to determine the conversion between assays. Net reclassification was determined after mathematical conversion. RESULTS: The Luminex assay was precise, with an intra- and interassay coefficient of variation 8.1% and 9.3%; showed modest agreement between 4 laboratories (R 0.96 to 0.99, P < 0.001); and correlated with known CXCL10 in a single- (n = 100 urines, R 0.94 to 0.98, P < 0.001) and multicenter cohort (n = 468 urines, R 0.92, P < 0.001) but the 2 assays were not equivalent by Passing-Bablok regression. Linear mixed-effects modeling demonstrated an intercept of -0.490 and coefficient of 1.028, showing Luminex CXCL10 are slightly higher than MSD CXCL10, but the agreement is close to 1.0. After conversion of the biopsy thresholds, the decision to biopsy would be changed for only 6% (5/85) patients showing acceptable reclassification. CONCLUSIONS: These data demonstrate this urine CXCL10 Luminex immunoassay is robust, reproducible, and accurate, indicating it can be readily translated into clinical HLA laboratories for serial posttransplant surveillance.


Subject(s)
Kidney Transplantation , Adult , Humans , Kidney Transplantation/adverse effects , Chemokine CXCL10 , Biomarkers , Interferon-gamma , Immunoassay , Graft Rejection/diagnosis
6.
BMC Gastroenterol ; 22(1): 34, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35078405

ABSTRACT

BACKGROUND: Transplantation offers the best survival for patients with end stage organ disease. Transplant of hepatitis C virus (HCV) nucleic acid test (NAT) positive organs into negative recipients is a novel strategy that can expand the donor pool. We aim to evaluate our centre's experience. METHODS: We preformed a retrospective review of anti-HCV NAT positive and negative organs into negative recipients transplanted over 27 months. Primary outcome was the success rate of eradication of HCV post-transplant. Secondary outcomes were rate of transmission of HCV, treatment adverse events, and graft failure. RESULTS: 33 anti-HCV positive organs were transplanted into negative recipients. 22 (66.7%) were NAT positive. Median recipients age was 49 years (interquartile range [IQR] 44.5-62.0) with the majority being males (57.6%). NAT positive organ transplantations included 16 kidneys, 3 livers, 1 kidney-pancreas, 1 liver-kidney, and 1 heart. The most common HCV genotype was 1a (59.1%). The median time to initiating therapy was 41.5 days. SVR12 was 100% in patients who finished therapy. There were no adverse events with therapy and no graft failure. CONCLUSIONS: Anti-HCV NAT positive organ transplantation into negative recipients is safe with excellent eradication rates and no significant adverse events or graft failure. This would expand donor pool to close the gap between supply and demand.


Subject(s)
Hepatitis C , Organ Transplantation , Canada , Hepacivirus/genetics , Hepatitis C/drug therapy , Humans , Male , Middle Aged , Organ Transplantation/adverse effects , Retrospective Studies
7.
Transplant Proc ; 53(5): 1570-1575, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33994184

ABSTRACT

BACKGROUND: The presence of intimal arteritis (v) in renal allograft biopsy specimens establishes the presence of acute T-cell mediated rejection (TCMR), Grade IIa-III, according to the Banff classification of rejection. The clinical significance of isolated v1 lesions (v1), characterized by arteritis alone, compared with lesions of arteritis with tubulointerstitial inflammation (i-t-v) has been controversial. METHODS: We performed a retrospective review of 280 patients undergoing renal transplantation between 2005 and 2015 who received a "for cause" transplant biopsy using the Banff 2013 classification. Patients with TCMR grade IIa (n = 83) were subdivided into groups with isolated v1 arteritis and i-t-v. Pre- and postoperative renal function, graft survival, and overall survival were evaluated in all patients. RESULTS: Donor and recipient demographics were similar between groups. One month following treatment of rejection, patients with v1 disease had superior recovery of glomerular filtration rate vs patients with i-t-v (P < .002). At a median follow-up of 41 months from transplant, death-censored graft survival was 92% vs 79% (P = .04), and overall survival was 98% vs 79% (P < .004) in the isolated v1 and i-t-v groups, respectively. CONCLUSION: Despite having identical Banff classification of TCMR IIa, our results indicate that graft survival in patients with isolated v1 rejection is superior to those with i-t-v. Following corroboration with data from other centers, modification of the Banff classification scheme should be considered.


Subject(s)
Arteritis/immunology , Graft Rejection/immunology , Kidney Transplantation/adverse effects , Nephritis, Interstitial/immunology , Postoperative Complications/immunology , Adult , Biopsy , Glomerular Filtration Rate , Graft Survival/immunology , Humans , Kidney/blood supply , Male , Middle Aged , Retrospective Studies , T-Lymphocytes/immunology , Transplants/blood supply
8.
Clin Transplant ; 35(7): e14322, 2021 07.
Article in English | MEDLINE | ID: mdl-33882151

ABSTRACT

BACKGROUND: Solid organ transplant (SOT) recipients are at risk of Pneumocystis pneumonia (PCP). PCP is associated with significant morbidity and mortality. The effect of acute T cell-mediated rejection (TCMR) on post-transplant PCP has not been determined yet. METHODS: In this case-control study, we estimated the risk of PCP following acute TCMR during a lookback period of 180 days. We also determined the effects of contributing factors such as CMV infection. RESULTS: We compared 15 SOT (8 kidney, 4 heart, 2 liver, and 1 kidney-pancreas) recipients with PCP with 60 matched recipients who did not develop PCP (control group) during the study period (December 2013 to February 2016). PCP occurred after a complete course of prophylaxis (ie, late-onset PCP) in 60% of patients. Patients with PCP frequently required intensive care unit (ICU) admission (73.3%). Post-transplant PCP was associated with considerable allograft loss (53.4%) and mortality (26.7%). In the 6-month lookback period, acute TCMR (OR: 13.1, 95% CI: 3.2, 53.2), and CMV infection (OR: 15.1,95% CI: 4.0, 53.2.1) were significantly associated with post-transplant PCP. CONCLUSIONS: Post-transplant PCP is associated with substantial risk of ICU admission, allograft failure, and mortality. Anti-Pneumocystis prophylaxis for at least 6 months following acute TCMR may reduce the risk.


Subject(s)
Organ Transplantation , Pneumocystis carinii , Pneumonia, Pneumocystis , Case-Control Studies , Graft Rejection/epidemiology , Graft Rejection/etiology , Humans , Organ Transplantation/adverse effects , Pneumonia, Pneumocystis/epidemiology , Pneumonia, Pneumocystis/etiology , Transplant Recipients
9.
Am J Transplant ; 21(10): 3268-3279, 2021 10.
Article in English | MEDLINE | ID: mdl-33784431

ABSTRACT

Inflammation posttransplant is directly linked to cell death programs including apoptosis and necrosis. Cell death leads to the release of cellular contents which can promote inflammation. Targeting of these pathways should be an effective strategy to prevent transplant rejection. Toll-like receptor 3 (TLR3) is emerging as a major endogenous sensor of inflammation. In this study, we assessed the role of TLR3 on cell death and transplant rejection. We showed that TLR3 is highly expressed on mouse microvascular endothelial cell (ECs) and the endothelium of cardiac grafts. We demonstrated that TLR3 interacting with dsRNA or self-RNA triggered apoptosis and necroptosis in ECs. Interestingly, TLR3-induced necroptosis led mitochondrial damage. Inhibition of the mitochondrial membrane permeability molecule Cyclophilin D prevented necroptosis in ECs. In vivo, endothelium damage and activities of caspase-3 and mixed lineage kinase domain-like protein were inhibited in TLR3-/- cardiac grafts compared with C57BL/6 grafts posttransplant (n = 5, p < .001). Importantly, TLR3-/- cardiac grafts had prolonged survival in allogeneic BALB/c mice (mean survival = 121 ± 67 vs. 31 ± 6 days of C57BL/6 grafts, n = 7, p = .002). In summary, our study suggests that TLR3 is an important cell death inducer in ECs and cardiac grafts and thus a potential therapeutic target in preventing cardiac transplant rejection.


Subject(s)
Heart Transplantation , Toll-Like Receptor 3 , Animals , Apoptosis , Cell Death , Heart Transplantation/adverse effects , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tissue Donors , Toll-Like Receptor 3/metabolism
10.
Clin Transplant ; 35(6): e14285, 2021 06.
Article in English | MEDLINE | ID: mdl-33713374

ABSTRACT

Renal transplant recipients remain at risk of delayed-onset cytomegalovirus (CMV) infection occurring beyond a complete course of prophylaxis. In this retrospective cohort, all 278 patients who received renal allografts from deceased donors from 2014 to 2016 were followed until September 1, 2019. We determined the effect of early-vs late-onset acute rejection (EAR vs LAR [ie, occurring beyond 12 months after transplantation]) on CMV infection and subsequently long-term allograft outcome. Median (IQR) duration of follow-up was 1186.0 (904.7-1531.2) days. Seventy patients including 49 patients with EAR and 21 with LAR received augmented immunosuppression. In the same interval, 40 patients developed CMV infection (36 patients beyond 90 days after transplantation [90%]). In logistic regression analysis, D+/R- CMV serostatus (OR: 5.5, 95% CI: 2.5-12.2) and LAR (OR: 7.9, 95% CI: 2.8-22.2) significantly increased the risk of CMV infection. In Cox proportional hazard model, delayed-onset CMV infection (HR: 2.51, 95% CI: 1.08-5.86) and LAR (HR: 5.46, 95% CI: 2.26-13.14) significantly increased the risk of allograft loss. Patients with LAR are at risk of late-onset CMV infection. Post-LAR, targeted prophylaxis may reduce the risk of CMV infection and subsequently allograft loss. Further studies are required to demonstrate the effect of targeted prophylaxis following LAR.


Subject(s)
Cytomegalovirus Infections , Kidney Transplantation , Allografts , Antiviral Agents/therapeutic use , Cytomegalovirus , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/etiology , Cytomegalovirus Infections/prevention & control , Graft Rejection/etiology , Graft Rejection/prevention & control , Humans , Kidney Transplantation/adverse effects , Retrospective Studies , Risk Factors
11.
Clin Transplant ; 34(12): e14108, 2020 12.
Article in English | MEDLINE | ID: mdl-33048378

ABSTRACT

Pneumocystis pneumonia (PCP) outbreaks may occur in solid organ transplant (SOT) patients. Transmissibility of Pneumocystis jirovecii among SOT and non-SOT patients has not been investigated. Ten SOT (ie, 4 heart, 4 kidney, 2 liver allograft recipients) and 11 non-SOT (ie, 7 HIV-infected, 3 hematologic malignancies, and 1 stem cell transplant) patients with PCP were admitted to London Health Sciences Center (LHSC) from October 2014 to August 2016. We investigated the course of illness and outcome of PCP in SOT and non-SOT patients. Post-transplant PCP was frequently an acute-onset disease (90% vs. 18.2%, p = .01) requiring ICU admission (70% vs. 20%, p = .03) and hemodialysis (60% vs. 0, p = .003). Mortality was more frequent in SOT patients (40% vs. 18.1%, p = .36). Multilocus sequence typing (MLST) demonstrated circulation of a single genotype of P. jirovecii among SOT patients. However, 8 different genotypes were detected from non-SOT patients. Reinstitution of prophylaxis successfully controlled post-transplant cluster until end of observation period in October 2019. No transmission was detected from non-SOT patients to SOT recipients. Detection of a single P. jirovecii genotype from all SOT recipients highlights the likelihood of nosocomial transmission. No source control method is recommended by current guidelines. Improvement of preventive strategies is required.


Subject(s)
Cross Infection , Healthcare-Associated Pneumonia , Pneumocystis carinii , Pneumonia, Pneumocystis , Allografts , Cross Infection/epidemiology , Cross Infection/etiology , Genotype , Humans , Multilocus Sequence Typing , Pneumocystis carinii/genetics , Pneumonia, Pneumocystis/diagnosis , Pneumonia, Pneumocystis/epidemiology , Pneumonia, Pneumocystis/etiology
12.
Clin Transplant ; 34(12): e14110, 2020 12.
Article in English | MEDLINE | ID: mdl-33053214

ABSTRACT

Human leukocyte antigen (HLA) class I presentation pathway plays a central role in natural killer (NK) cell and cytotoxic T-cell activities against BK polyomavirus (BKPyV) DNAemia. We determined the risk of sustained BKPyV DNAemia in 175 consecutive renal transplant recipients considering the simultaneous effect of donor/recipient HLA class I antigens and pre- or post-transplant variables. Median (IQR) age was 53 (44-64) years, and 37% of patients were female. 40 patients (22.9%) developed sustained BKPyV DNAemia [median (IQR) viral load: 9740 (4350-17 125) copies/ml]. In the Cox proportional hazard analysis, HLA-A1 (HR: 3.06, 95% CI: 1.51-6.17) and HLA-B35-Cw4 (HR: 4.63, 95% CI: 2.12-10.14) significantly increased the risk of sustained BKPyV DNAemia, while 2 HLA-C mismatches provided a marginally protective effect (HR: 0.32, 95% CI: 0.10-0.98). HLA-Cw4 is a ligand for NK cell inhibitory receptor, and HLA-B35 is in strong linkage disequilibrium with the HLA-Cw4 allele. The association between HLA-B35-Cw4 expression and sustained BKPyV DNAemia supports the important role of cytotoxic T cells and NK cells that would normally control BKPyV activation through engagement with immunoglobulin-like killer receptors (KIRs). Further studies are required to investigate the effect of HLA-C alleles along with NK cell activity against BKPyV DNAemia.


Subject(s)
BK Virus , Kidney Transplantation , Polyomavirus Infections , Adult , BK Virus/genetics , Female , HLA-A1 Antigen , Humans , Kidney Transplantation/adverse effects , Male , Middle Aged , Polyomavirus Infections/etiology , Transplant Recipients
13.
Can J Kidney Health Dis ; 7: 2054358119898552, 2020.
Article in English | MEDLINE | ID: mdl-32047642

ABSTRACT

BACKGROUND: In Ontario, kidney transplants have risen by 4% annually in recent years. An understanding of how this will affect the future annual number of posttransplant follow-up visits informs how to organize and deliver care. OBJECTIVE: We projected the required number of annual posttransplant follow-up nephrology visits to inform posttransplant care delivery. DESIGN: Population-based retrospective cohort study. SETTING: Linked databases from Ontario, Canada (population 14 million). PATIENTS: Incident kidney transplant recipients from years 2008 to 2013. MEASUREMENTS: Frequency, distance traveled, and current and projected visits for posttransplant follow-up. METHODS: Assuming a graft survival of 13 years and using the mean number of posttransplant clinic visits in years 1, 2, and 3, we forecasted the number of clinic visits needed in the year 2027. RESULTS: Using data from 2443 recipients, the mean (SD) number of clinic visits per recipient was 14.0 (9.2) in the first year after transplant, and 3.9 (6.2) and 3.0 (5.3) in the second and third year, respectively. If transplant rates rise by 4% per year until 2027, the estimated annual visits number will increase from 30 622 to 43 948. The median (25th, 75th percentile) distance between transplant center and patient's home was 30 (13, 65) km. The median round-trip travel distance for these visits in the first year after transplantation was 603 km per recipient, and median driving cost was Can$344 (2017). LIMITATIONS: Regarding patient expense, limitations include that distances traveled were calculated orthodromically, and we did not account for patient cost of follow-up beyond that of vehicular travel. Regarding follow-up projections, limitations include the assumption that graft life span will not change, follow-up patterns do not differ between donor kidney type, and we did not survey stakeholders as to their preferred method of follow-up. CONCLUSION: We quantified the increase in posttransplant visits when regional annual rates of transplantation rise. Strategies recognizing the burden of these visits may enhance patient-centered care, as it is unclear how some patients manage costs, nor how the current health care system will manage the demand.


CONTEXTE: Au cours des dernières années, le nombre de transplantations rénales a augmenté de 4 % annuellement en Ontario. Comprendre l'impact qu'aura cette augmentation sur le nombre annuel de visites de suivi post-transplantation dans les années à venir permettra d'organiser et de prodiguer les soins aux patients. OBJECTIF: Prévoir le nombre de visites de suivi post-transplantation qui seront requises dans le futur, de façon à orienter la prestation des soins. TYPE D'ÉTUDE: Étude de cohorte rétrospective basée sur une population. CADRE: Les bases de données couplées de l'Ontario, au Canada (population: 14 millions). SUJETS: Les receveurs d'une greffe rénale entre 2008 et 2013. MESURES: La fréquence des visites post-transplantation, la distance à parcourir pour s'y rendre, de même que leur nombre actuel et projeté. MÉTHODOLOGIE: Nous avons estimé le nombre de visites cliniques qui seront nécessaires en 2027 à partir d'une survie du greffon estimée à 13 ans et du nombre moyen de visites pour les années 1, 2 et 3 post-transplantation. RÉSULTATS: À partir des données de 2 443 receveurs, nous avons établi le nombre moyen (ET) de visites cliniques par receveur à 14,0 (9,2) pour la première année post-transplantation, à 3,9 (6,2) pour l'an 2 et à 3,0 (5,3) pour l'an 3. Si le nombre de greffes poursuit sa croissance au rythme de 4 % par an jusqu'en 2027, on estime que le nombre annuel de visites passera de 30 622 à 43 948. La distance médiane (25e, 75e percentile) entre le centre de greffe et la résidence du patient était de 30 km (13, 65). Dans l'année suivant la greffe, chaque patient avait parcouru une distance médiane totale (aller-retour) de 603 km et avait dépensé 344 $ (coût médian en dollars canadiens en 2017) pour ces déplacements. LIMITES: Les distances parcourues par les patients n'ont été calculées que de manière orthodromique et seules les dépenses liées aux déplacements en voiture ont été employées pour calculer les coûts aux patients. Les prévisions pour 2027 sont établies en tenant compte d'une survie du greffon qui demeurera stable, d'un schéma de suivi qui ne différera pas selon le type de donneur et du fait que nous n'avons pas questionné les intervenants quant à leur méthode de suivi préférée. CONCLUSION: Nous avons quantifié la hausse du nombre de visites post-transplantation en fonction de l'augmentation des taux de greffes annuels régionaux. Des stratégies reconnaissant le fardeau imposé par ces visites ont le potentiel d'améliorer les soins axés sur les patients puisqu'on ignore comment certains patients gèrent les coûts associés et comment le système de santé gèrera la demande.

14.
BMJ Open ; 9(4): e024908, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975673

ABSTRACT

INTRODUCTION: Subclinical inflammation is an important predictor of death-censored graft loss, and its treatment has been shown to improve graft outcomes. Urine CXCL10 outperforms standard post-transplant surveillance in observational studies, by detecting subclinical rejection and early clinical rejection before graft functional decline in kidney transplant recipients. METHODS AND ANALYSIS: This is a phase ii/iii multicentre, international randomised controlled parallel group trial to determine if the early treatment of rejection, as detected by urine CXCL10, will improve kidney allograft outcomes. Incident adult kidney transplant patients (n~420) will be enrolled to undergo routine urine CXCL10 monitoring postkidney transplant. Patients at high risk of rejection, defined as confirmed elevated urine CXCL10 level, will be randomised 1:1 stratified by centre (n=250). The intervention arm (n=125) will undergo a study biopsy to check for subclinical rejection and biopsy-proven rejection will be treated per protocol. The control arm (n=125) will undergo routine post-transplant monitoring. The primary outcome at 12 months is a composite of death-censored graft loss, clinical biopsy-proven acute rejection, de novo donor-specific antibody, inflammation in areas of interstitial fibrosis and tubular atrophy (Banff i-IFTA, chronic active T-cell mediated rejection) and subclinical tubulitis on 12-month surveillance biopsy. The secondary outcomes include decline of graft function, microvascular inflammation at 12 months, development of IFTA at 12 months, days from transplantation to clinical biopsy-proven rejection, albuminuria, EuroQol five-dimension five-level instrument, cost-effectiveness analysis of the urine CXCL10 monitoring strategy and the urine CXCL10 kinetics in response to rejection therapy. ETHICS AND DISSEMINATION: The study has been approved by the University of Manitoba Health Research Ethics Board (HS20861, B2017:076) and the local research ethics boards of participating centres. Recruitment commenced in March 2018 and results are expected to be published in 2023. De-identified data may be shared with other researchers according to international guidelines (International Committee of Medical Journal Editors [ICJME]). TRIAL REGISTRATION NUMBER: NCT03206801; Pre-results.


Subject(s)
Chemokine CXCL10/urine , Delayed Graft Function/urine , Graft Rejection/urine , Kidney Transplantation , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Adult , Biomarkers/urine , Female , Health Surveys , Humans , Kidney Transplantation/adverse effects , Male , Middle Aged , Predictive Value of Tests
15.
Hum Immunol ; 80(7): 487-492, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30904438

ABSTRACT

Despite implementation of virtual crossmatches, flow cytometry crossmatches (FCXM) are still used by many transplant centers to determine immunological risk before kidney transplantation. To determine if common profiles of patients prone to false positive FCXM exist, we examined the demographics and native diseases of kidney patients tested with autologous FCXM (n = 480). Improvements to FCXM and cell isolation methods significantly reduced the positive rate from 15.1% to 5.3%. Patients with native diseases considered 'immunological' (vasculitis, lupus, IgA nephropathy) had more positive autologous FCXM (OR = 3.36, p = 0.003) vs. patients with all other diseases. Patients who were tested using our updated method (n = 321) still showed that these immunological diseases were a significant predictor for positive autologous FCXM (OR = 4.79, p = 0.006). Interestingly, patients on peritoneal dialysis (PD) also had significantly more positive autologous FCXM than patients on hemodialysis or waiting for pre-emptive kidney transplants (OR = 3.27, p = 0.02). These findings were confirmed in patients who had false positive allogeneic FCXM. Twenty of 24 (83.3%) patients with false positive allogeneic FCXM tested with updated method either had immunological diseases originally or were on PD. Our findings are helpful when interpreting an unexpected positive FCXM, especially for transplantation from deceased donors.


Subject(s)
Autoantibodies/immunology , Flow Cytometry/methods , Glomerulonephritis, IGA/immunology , Histocompatibility Testing/methods , Lupus Erythematosus, Systemic/immunology , Peritoneal Dialysis , Autoantigens/immunology , Cohort Studies , Female , Graft Rejection/immunology , HLA Antigens/immunology , Humans , Kidney Transplantation , Linear Models , Logistic Models , Male , Middle Aged , Tissue Donors
16.
Plant Cell Rep ; 38(3): 391-401, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30659328

ABSTRACT

KEY MESSAGE: We demonstrate for the first time that a fully bioactive human IL-37, a newly discovered cytokine acting as a fundamental inhibitor of innate immunity, can be recombinantly produced in plant cells. Interleukin 37 (IL-37), a newly discovered member of the interleukin (IL)-1 family of cytokines, plays a pivotal role in limiting innate inflammation and suppressing acquired immune responses, thus holding high potential for treating a wide array of human inflammatory and autoimmune disorders. In this study, we have developed transgenic plants as a novel expression platform for production of human IL-37 (IL-37). Plant transformation vectors synthesizing various forms of the b isoform of IL-37, including an unprocessed full-length precursor form (proIL-37b), a mature form (matIL-37b) and an IL-37 fusion protein in which IL-37b was fused to soybean agglutinin (SBA-IL-37b), have been constructed and introduced into tobacco plants. The expression of all forms of IL-37b was driven by a strong constitutive 35S promoter. Transgenic tobacco plants were generated with each of these constructs. Depending on the form of IL-37b being produced, the expression level of proIL-37b reached approximately 1% of TSP, while matIL-37b expression was substantially lower (0.01% TSP). Fusion to SBA substantially increased the expression of matIL-37b, with the expression level of fusion protein accounting for 1% of TSP. Functional analysis using a cell-based in vitro assay showed that plant-made matIL-37b and proIL-37b are both biologically active, but plant-made matIL-37b exhibited significantly greater biological activity than proIL-37b. These results demonstrate that plants have great potential of being a green bioreactor for low-cost, large-scale production of biologically active IL-37.


Subject(s)
Interleukin-1/metabolism , Cytokines/metabolism , Humans , Immunity, Innate/genetics , Immunity, Innate/physiology , Interleukin-1/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Nicotiana/genetics , Nicotiana/metabolism
17.
Nephrology (Carlton) ; 24(6): 661-669, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30175514

ABSTRACT

BACKGROUND: Ischaemia-reperfusion injury (IRI) is associated with programmed cell death that promotes inflammation and organ dysfunction. Necroptosis is mediated by members of receptor interacting protein kinases (RIPK1/3). Inhibition of RIPK1/3 provides a pro-survival benefit in kidney IRI. Caspase-8 initiates apoptosis and contributes to IRI. We studied whether inhibiting both RIPK3 and caspase-8 would provide an additional benefit in kidney IRI. METHODS: A clamp was applied to the left kidney pedicle for 45 min followed by right kidney nephrectomy. Kidney and serum from wild type, RIPK3-/- , and RIPK3-/- caspase-8-/- double knockout (DKO) mice were collected post-IRI for assessment of injury. Tubular epithelial cells (TEC) isolated from wild type, RIPK3-/- , and DKO mice were treated with interferons-γ and interleukin-1ß to induce apoptotic death. RESULTS: Kidney IRI of DKO mice did not show improvement over RIPK3-/- mice. We have found that DKO triggered 'intrinsic' apoptosis in TEC in response to interleukin-1ß and interferons-γ. Up-regulation of the B-cell lymphoma 2 (Bcl-2)-associated death promoter, the Bcl-2-homologous antagonist killer and Bcl-2-associated X protein and enhanced activation of caspase-3 and 9 were found in DKO TEC. TEC infected with Murine cytomegalovirus that encodes multiple cell death inhibitors resist to death. CONCLUSION: We show that the deletion of both RIPK3 and caspase-8 does not provide additive benefit in IRI or TEC death and may enhance injury by up-regulation of intrinsic apoptosis. This suggests blocking multiple death pathways may be required for the prevention of kidney IRI clinically.


Subject(s)
Apoptosis , Caspase 8/metabolism , Epithelial Cells/enzymology , Kidney Diseases/enzymology , Kidney Tubules/enzymology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Reperfusion Injury/enzymology , Animals , Apoptosis/drug effects , Caspase 8/genetics , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction
18.
Am J Transplant ; 19(3): 686-698, 2019 03.
Article in English | MEDLINE | ID: mdl-30203531

ABSTRACT

Transplantation is invariably associated with programmed cell death including apoptosis and necrosis, resulting in delayed graft function and organ rejection. We have demonstrated the contribution of necroptosis to mouse microvascular endothelial cell (MVEC) death and transplant rejection. Organ injury results in the opening of mitochondrial permeability transition pores (mPTPs), which can trigger apoptotic molecules release that ultimately results in cell death. The effect of mPTPs in the necroptotic pathway remains controversial; importantly, their role in transplant rejection is not clear. In this study, tumor necrosis factor-α triggered MVECs to undergo receptor-interacting protein kinase family (RIPK1/3)-dependent necroptosis. Interestingly, inhibition of mPTP opening could also inhibit necroptotic cell death. Cyclophilin-D (Cyp-D) is a key regulator of the mPTPs. Both inhibition and deficiency of Cyp-D protected MVECs from necroptosis (n = 3, P < .00001). Additionally, inhibition of Cyp-D attenuated RIPK3-downstream mixed-lineage kinase domain-like protein phosphorylation. In vivo, Cyp-D-deficient cardiac grafts showed prolonged survival in allogeneic BALB/c mice posttransplant compared with wild-type grafts (n = 7, P < .0001). Our study results suggest that the mPTPs may be important mechanistic mediators of necroptosis in cardiac grafts. There is therapeutic potential in targeting cell death via inhibition of the mPTP-regulating molecule Cyp-D to prevent cardiac graft rejection.


Subject(s)
Cell Membrane Permeability , Endothelial Cells/pathology , Graft Rejection/etiology , Heart Transplantation/adverse effects , Mitochondria/pathology , Necroptosis , Peptidyl-Prolyl Isomerase F/metabolism , Allografts , Animals , Peptidyl-Prolyl Isomerase F/genetics , Endothelial Cells/immunology , Endothelial Cells/metabolism , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/immunology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tissue Donors , Tumor Necrosis Factor-alpha/pharmacology
19.
Am J Transplant ; 19(6): 1730-1744, 2019 06.
Article in English | MEDLINE | ID: mdl-30582281

ABSTRACT

Targeting the renin-angiotensin system and optimizing tacrolimus exposure are both postulated to improve outcomes in renal transplant recipients (RTRs) by preventing interstitial fibrosis/tubular atrophy (IF/TA). In this multicenter, prospective, open-label controlled trial, adult de novo RTRs were randomized in a 2 × 2 design to low- vs standard-dose (LOW vs STD) prolonged-release tacrolimus and to angiotensin-converting enzyme inhibitors/angiotensin II receptor 1 blockers (ACEi/ARBs) vs other antihypertensive therapy (OAHT). There were 2 coprimary endpoints: the prevalence of IF/TA at month 6 and at month 24. IF/TA prevalence was similar for LOW vs STD tacrolimus at month 6 (36.8% vs 39.5%; P = .80) and ACEi/ARBs vs OAHT at month 24 (54.8% vs 58.2%; P = .33). IF/TA progression decreased significantly with LOW vs STD tacrolimus at month 24 (mean [SD] change, +0.42 [1.477] vs +1.10 [1.577]; P = .0039). Across the 4 treatment groups, LOW + ACEi/ARB patients exhibited the lowest mean IF/TA change and, compared with LOW + OAHT patients, experienced significantly delayed time to first T cell-mediated rejection. Renal function was stable from month 1 to month 24 in all treatment groups. No unexpected safety findings were detected. Coupled with LOW tacrolimus dosing, ACEi/ARBs appear to reduce IF/TA progression and delay rejection relative to reduced tacrolimus exposure without renin-angiotensin system blockade. ClinicalTrials.gov identifier: NCT00933231.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Kidney Transplantation/methods , Tacrolimus/administration & dosage , Adult , Allografts , Atrophy , Delayed-Action Preparations , Drug Therapy, Combination , Female , Fibrosis , Graft Rejection/etiology , Graft Rejection/immunology , Humans , Immunosuppressive Agents/administration & dosage , Kidney/pathology , Kidney/physiopathology , Kidney Transplantation/adverse effects , Male , Middle Aged , Polyomavirus Infections/etiology , Prognosis , Prospective Studies , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Virus Activation
20.
J Clin Invest ; 128(7): 2894-2913, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29863495

ABSTRACT

Radiographic contrast agents cause acute kidney injury (AKI), yet the underlying pathogenesis is poorly understood. Nod-like receptor pyrin containing 3-deficient (Nlrp3-deficient) mice displayed reduced epithelial cell injury and inflammation in the kidney in a model of contrast-induced AKI (CI-AKI). Unexpectedly, contrast agents directly induced tubular epithelial cell death in vitro that was not dependent on Nlrp3. Rather, contrast agents activated the canonical Nlrp3 inflammasome in macrophages. Intravital microscopy revealed diatrizoate (DTA) uptake within minutes in perivascular CX3CR1+ resident phagocytes in the kidney. Following rapid filtration into the tubular luminal space, DTA was reabsorbed and concentrated in tubular epithelial cells via the brush border enzyme dipeptidase-1 in volume-depleted but not euvolemic mice. LysM-GFP+ macrophages recruited to the kidney interstitial space ingested contrast material transported from the urine via direct interactions with tubules. CI-AKI was dependent on resident renal phagocytes, IL-1, leukocyte recruitment, and dipeptidase-1. Levels of the inflammasome-related urinary biomarkers IL-18 and caspase-1 were increased immediately following contrast administration in patients undergoing coronary angiography, consistent with the acute renal effects observed in mice. Taken together, these data show that CI-AKI is a multistep process that involves immune surveillance by resident and infiltrating renal phagocytes, Nlrp3-dependent inflammation, and the tubular reabsorption of contrast via dipeptidase-1.


Subject(s)
Acute Kidney Injury/etiology , Contrast Media/adverse effects , Dipeptidases/metabolism , Immunologic Surveillance , Kidney/enzymology , Kidney/immunology , Acute Kidney Injury/immunology , Acute Kidney Injury/metabolism , Animals , Contrast Media/pharmacokinetics , Disease Models, Animal , GPI-Linked Proteins/metabolism , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Kidney/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Phagocytes/immunology , Phagocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...