1
|
Tavee J. Current concepts in long COVID-19 brain fog and postural orthostatic tachycardia syndrome. Ann Allergy Asthma Immunol 2024; 133:522-530. [PMID: 39154907 DOI: 10.1016/j.anai.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024]
Abstract
Neurologic complications of long COVID-19 syndrome are one of the leading causes of global disability. In particular, post-COVID-19 cognitive dysfunction and dysautonomia in the form of postural orthostatic tachycardia syndrome (POTS) markedly affect patient quality of life and ability to return to work. The underlying pathophysiology of post-COVID-19 neurologic complications is unknown but is likely multifactorial with immune dysregulation and microvascular dysfunction playing central roles. Specific pathogenic factors with supportive evidence to date include cytokine-mediated inflammation, autoantibodies, immune exhaustion, disruption of the renin-angiotensin system, reduced serotonin levels, and microglial activation. The prevalence of post-COVID-19 cognitive dysfunction ranges from 10% to 88% and is affected by viral variant and hospitalization status among other factors, whereas that of long COVID-19 POTS is unknown due to referral bias and varying definitions. Treatment is largely supportive and often incorporates combined modalities. Marginal benefits with cognitive behavioral therapy, hyperbaric oxygen therapy, and supplements have been found for post-COVID-19 brain fog, whereas established POTS therapies aimed at improving venous return and reducing heart rate may reduce symptoms of long COVID-19 POTS. Although significant recovery has been noted for many cases of post-COVID-19 brain fog and POTS, prospective studies have revealed evidence of persistent symptoms and neurologic deficits a year after infection in some patients. Further studies that provide insight into the underlying pathophysiology of long COVID-19 are needed for development of target directed therapy.
Collapse
Affiliation(s)
- Jinny Tavee
- Division of Neurology, National Jewish Health, Denver, Colorado.
| |
Collapse
|
2
|
Kermond RF, Kim S, Mackie F, Hahn D, Carroll RP, Sharma A, Durkan AM. Effect of angiotensin II type 1 receptor antibodies on graft function and survival in paediatric kidney transplant recipients. HLA 2024; 104:e15649. [PMID: 39247998 DOI: 10.1111/tan.15649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/25/2024] [Accepted: 08/03/2024] [Indexed: 09/10/2024]
Abstract
HLA donor specific antibodies (DSA) are implicated in antibody-mediated rejection (AMR), graft dysfunction and failure in kidney transplant (KT) recipients. Non-HLA antibodies including angiotensin II type 1 receptor (AT1R) may also play a role in AMR, impact graft function and survival. Data is limited in paediatric KT cohorts. We aimed to assess the prevalence and effect of pre-transplant AT1R antibodies on rejection, graft function and survival in paediatric KT recipients. This was a retrospective cohort study conducted across two paediatric centres including KT recipients with a pre-transplant AT1R antibody level. Outcomes included rejection, de novo DSA formation, graft function, failure, proteinuria and hypertension. Of 71 individuals, 72% recorded a positive pre-transplant AT1R Ab level (≥17 U/mL). Over a median follow-up of 4.7 years, AT1R Ab positivity demonstrated a trend towards increased risk of rejection however was not statistically significant (HR 3.45, 95% CI 0.97-12.35, p-value 0.06). Sensitivity analysis with AT1R Ab levels of ≥25 U/mL (HR 2.05 95% CI 0.78-5.39, p-value 0.14) and ≥40 U/mL (HR 1.32, CI 95% 0.55-3.17, p-value 0.53) validated this. De novo DSA formation occurred more frequently with AT1R Ab positivity (41% vs. 20%, p-value 0.9). AT1R Ab was not associated with hypertension, proteinuria, graft failure or dysfunction. In conclusion, this cohort study demonstrated a high prevalence of pre-transplant AT1R Ab positivity (72%). AT1R Ab positivity demonstrated a trend towards increased risk of rejection and de novo DSA formation however did not meet statistical significance. There was no association between AT1R Ab and hypertension, proteinuria, graft failure or dysfunction.
Collapse
Affiliation(s)
- R F Kermond
- Department of Paediatric Nephrology, Women's and Children's Hospital, Adelaide, Australia
- School of Women's and Children's Health, University of New South Wales, Kensington, Australia
| | - S Kim
- Department of Paediatric Nephrology, The Children's Hospital Westmead, Sydney, Australia
- Centre for Kidney research, Children's Hospital Westmead, Sydney, Australia
- School of Public Health, Sydney University, Sydney, Australia
| | - F Mackie
- School of Women's and Children's Health, University of New South Wales, Kensington, Australia
- Department of Paediatric Nephrology, Sydney Children's Hospital, Sydney, Australia
| | - D Hahn
- Department of Paediatric Nephrology, The Children's Hospital Westmead, Sydney, Australia
| | - R P Carroll
- South Australian Transplantation Immunogenetic Laboratory, Australian Red Cross Lifeblood, Adelaide, Australia
- Department of Health Sciences, University of South Australia, Adelaide, Australia
| | - A Sharma
- Department of Nephrology, Westmead Hospital, Sydney, Australia
| | - A M Durkan
- Department of Paediatric Nephrology, The Children's Hospital Westmead, Sydney, Australia
- School of Paediatrics and Child Health, University of Sydney, Australia
| |
Collapse
|
3
|
Botoni FA, Lambertucci JR, Santos RAS, Müller J, Talvani A, Wallukat G. Functional autoantibodies against G protein-coupled receptors in hepatic and pulmonary hypertensions in human schistosomiasis. Front Immunol 2024; 15:1404384. [PMID: 38953035 PMCID: PMC11216020 DOI: 10.3389/fimmu.2024.1404384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/23/2024] [Indexed: 07/03/2024] Open
Abstract
Introduction Schistosomiasis (SM) is a parasitic disease caused by Schistosoma mansoni. SM causes chronic inflammation induced by parasitic eggs, with collagen/fibrosis deposition in the granuloma process in the liver, spleen, central nervous system, kidneys, and lungs. Pulmonary arterial hypertension (PAH) is a clinical manifestation characterized by high pressure in the pulmonary circulation and right ventricular overload. This study investigated the production of functional autoantibodies (fAABs) against the second loop of the G-protein-coupled receptor (GPCR) in the presence of hepatic and PAH forms of human SM. Methods Uninfected and infected individuals presenting acute and chronic manifestations (e.g., hepatointestinal, hepato-splenic without PAH, and hepato-splenic with PAH) of SM were clinically evaluated and their blood was collected to identify fAABs/GPCRs capable of recognizing endothelin 1, angiotensin II, and a-1 adrenergic receptor. Human serum was analyzed in rat cardiomyocytes cultured in the presence of the receptor antagonists urapidil, losartan, and BQ123. Results The fAABs/GPCRs from chronic hepatic and PAH SM individuals, but not from acute SM individuals, recognized the three receptors. In the presence of the antagonists, there was a reduction in beating rate changes in cultured cardiomyocytes. In addition, binding sites on the extracellular domain functionality of fAABs were identified, and IgG1 and/or IgG3 antibodies were found to be related to fAABs. Conclusion Our data suggest that fAABs against GPCR play an essential role in vascular activity in chronic SM (hepatic and PAH) and might be involved in the development of hypertensive forms of SM.
Collapse
Affiliation(s)
- Fernando Antonio Botoni
- Postgraduate Program in Infectiology and Tropical Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Internal Medicine Department, School of Medicine Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Fundação Hospitalar do Estado de Minas Gerais - FHEMIG, Belo Horizonte, Brazil
| | - José Roberto Lambertucci
- Postgraduate Program in Infectiology and Tropical Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Internal Medicine Department, School of Medicine Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Postgraduate Program in Health and Nutrition, School of Nutrition, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and Biophysics of the Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Andre Talvani
- Postgraduate Program in Infectiology and Tropical Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Postgraduate Program in Health and Nutrition, School of Nutrition, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
- Department of Biological Sciences, Universidade Federal Ouro Preto, Ouro Preto, Brazil
| | - Gerd Wallukat
- Berlin Cures GmbH, Berlin, Germany
- Max-Delbrück Centrum für Molekulare Medizin, Berlin, Germany
| |
Collapse
|
4
|
Schmidt M, Hébert S, Wallukat G, Ponader R, Krickau T, Galiano M, Reutter H, Woelfle J, Agaimy A, Mardin C, Hoerning A, Hohberger B. "Multisystem Inflammatory Syndrome in Children"-Like Disease after COVID-19 Vaccination (MIS-V) with Potential Significance of Functional Active Autoantibodies Targeting G-Protein-Coupled Receptors (GPCR-fAAb) for Pathophysiology and Therapy. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1836. [PMID: 38136038 PMCID: PMC10741397 DOI: 10.3390/children10121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/28/2023] [Accepted: 10/28/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND An infection with SARS-CoV-2 can trigger a systemic disorder by pathological autoimmune processes. A certain type of this dysregulation is known as Multisystemic inflammatory syndrome in children (MIS-C). However, similar symptoms may occur and have been described as Multisystemic inflammatory syndrome after SARS-CoV-2 Vaccination (MIS-V) following vaccination against SARS-CoV-2. We report the case of a 12-year-old boy who was identified with MIS-C symptoms without previous SARS-CoV-2 infection after receiving two doses of the Pfizer-BioNTech COVID-19 vaccine approximately one month prior to the onset of symptoms. He showed polyserositis, severe gastrointestinal symptoms and, consequently, a manifestation of a multiorgan failure. IgG antibodies against spike proteins of SARS-CoV-2 were detected, indicating a successful vaccination, while SARS-CoV-2 Nucleocapsid protein antibodies and SARS-CoV-2 PCR were not detected. Several functional, active autoantibodies against G-protein-coupled receptors (GPCR-fAAb), previously associated with Long COVID disease, were detected in a cardiomyocyte bioassay. Immunosuppression with steroids was initiated. Due to side effects, treatment with steroids and later interleukin 1 receptor antagonists had to be terminated. Instead, immunoadsorption was performed and continued with tacrolimus and mycophenolic acid therapy, leading to improvement and discharge after 79 days. GPCR-fAAb decreased during therapy and remained negative after clinical curing and under continued immunosuppressive therapy with tacrolimus and mycophenolic acid. Follow-up of the patient showed him in good condition after one year. CONCLUSIONS Infection with SARS-CoV-2 shows a broad and severe variety of symptoms, partly due to autoimmune dysregulation, which, in some instances, can lead to multiorgan failure. Despite its rarity, post-vaccine MIS-C-like disease may develop into a serious condition triggered by autoimmune dysregulation. The evidence of circulating GPCR-fAAb and their disappearance after therapy suggests a link of GPCR-fAAb to the clinical manifestations. Thus, we hypothesize a potential role of GPCR-fAAb in pathophysiology and their potential importance for the therapy of MIS-C or MIS-V. However, this observation needs further investigation to prove a causative correlation.
Collapse
Affiliation(s)
- Marius Schmidt
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Steven Hébert
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | | | - Rolf Ponader
- Department of Pediatrics and Adolescent Medicine, 95032 Hof, Germany
| | - Tobias Krickau
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Matthias Galiano
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Heiko Reutter
- Division of Neonatology and Pediatric Intensive Care, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Abbas Agaimy
- Department of Pathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Christian Mardin
- Department of Ophthalmology, University Hospital Erlangen, 90766 Erlangen, Germany
| | - André Hoerning
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
- German Center for Immunotherapy, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, University Hospital Erlangen, 90766 Erlangen, Germany
- German Center for Immunotherapy, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany
| |
Collapse
|
5
|
Hébert S, Schmidt M, Topf G, Rieger D, Klinge J, Vermehren J, Fusch C, Grillhösl C, Schroth M, Toni I, Reutter H, Morhart P, Hanslik G, Mulzer L, Woelfle J, Hohberger B, Hoerning A. "Multisystem Inflammatory Syndrome in Children" (MIS-C) after COVID-19 Infection in the Metropolitan Area of Nuremberg-Erlangen, Germany-Expectations and Results of a Two-Year Period. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1363. [PMID: 37628362 PMCID: PMC10453116 DOI: 10.3390/children10081363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Multisystemic Inflammatory Syndrome in children (MIS-C) is a rare autoimmune disorder occurring after a latency period following acute SARS-CoV-2 infection. The therapeutic regime of MIS-C is adapted to the therapy of the Kawasaki disease, as clinical symptoms are similar. Since the Kawasaki disease can potentially result in severe symptoms, which may even affect long-term health, it is essential to gain further knowledge about MIS-C. Thus, we aimed to investigate the incidence, symptoms, therapeutical procedure and outcome of MIS-C patients in the metropolitan area of Nuremberg-Erlangen during the SARS-CoV2 pandemic. MATERIAL AND METHODS Retrospective analysis of clinical charts of MIS-C patients was carried out at three children's hospitals covering the medical care of the metropolitan area of Nuremberg-Erlangen in Germany. Demographic characteristics and symptoms at first visit, their clinical course, therapeutic regime and outcome were recorded within the time period January 2021-December 2022. RESULTS Analysis of 10 patients (5 male, 5 female) with MIS-C resulting in an incidence of 2.14/100.000 children. The median time between COVID-19 infection and admission to hospital was 5 weeks. The median age was 7 years. Symptoms comprised fever (100%), rash (70%), bilateral non-purulent conjunctivitis (70%) and urticaria (20%). At the time of presentation, diagnosis-defining inflammation parameters were increased and the range for C-reactive protein was 4.13 mg/dL to 28 mg/dL, with a median of 24.7 mg/dL. Procalcitonin was initially determined in six patients (1.92 ng/mL to 21.5 ng/mL) with a median value of 5.5 pg/mL. Two patients displayed leukocytosis and two displayed leukopenia. None of the patients presented coronary pathologies. Nine of the ten patients received intravenous immunoglobulin (IVIG) therapy. In addition, patients received intravenous steroids (80%) and acetylsalicylic acid (80%). CONCLUSION SARS-CoV virus may rarely exert multiorgan manifestations due to hyperinflammatory immunological processes. Within two years of the COVID-19 pandemic, we identified ten patients with COVID-induced MIS-C in the metropolitan area Nuremberg-Erlangen. In the description of the patient collective, we can confirm that MIS-C is distinguished from the Kawasaki disease by the lack of coronary manifestations. Interestingly, although having monitored all pediatric facilities in the investigated area, we find lower incidences of MIS-C compared to findings in the literature. In conclusion, an overestimation of incidences in the upcoming MIS-C during the pandemic needs to be considered.
Collapse
Affiliation(s)
- Steven Hébert
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Marius Schmidt
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Georg Topf
- Hospital for Children and Adolescents Furth, 90766 Furth, Germany
| | - Daniel Rieger
- Hospital for Children and Adolescents Furth, 90766 Furth, Germany
| | - Jens Klinge
- Hospital for Children and Adolescents Furth, 90766 Furth, Germany
| | - Jan Vermehren
- Pediatrics—Children’s Department Nuremberg Hospital South, 90471 Nuremberg, Germany
| | - Christoph Fusch
- Pediatrics—Children’s Department Nuremberg Hospital South, 90471 Nuremberg, Germany
| | - Christian Grillhösl
- Cnopf Children’s Hospital, Diakoneo Klinikum Hallerwiese Nuremberg, 90419 Nuremberg, Germany
| | - Michael Schroth
- Cnopf Children’s Hospital, Diakoneo Klinikum Hallerwiese Nuremberg, 90419 Nuremberg, Germany
| | - Irmgard Toni
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Heiko Reutter
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Patrick Morhart
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Gregor Hanslik
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Linda Mulzer
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, University Hospital Erlangen, 90766 Erlangen, Germany
| | - André Hoerning
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
6
|
Subburayalu J. Immune surveillance and humoral immune responses in kidney transplantation - A look back at T follicular helper cells. Front Immunol 2023; 14:1114842. [PMID: 37503334 PMCID: PMC10368994 DOI: 10.3389/fimmu.2023.1114842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/22/2023] [Indexed: 07/29/2023] Open
Abstract
T follicular helper cells comprise a specialized, heterogeneous subset of immune-competent T helper cells capable of influencing B cell responses in lymphoid tissues. In physiology, for example in response to microbial challenges or vaccination, this interaction chiefly results in the production of protecting antibodies and humoral memory. In the context of kidney transplantation, however, immune surveillance provided by T follicular helper cells can take a life of its own despite matching of human leukocyte antigens and employing the latest immunosuppressive regiments. This puts kidney transplant recipients at risk of subclinical and clinical rejection episodes with a potential risk for allograft loss. In this review, the current understanding of immune surveillance provided by T follicular helper cells is briefly described in physiological responses to contrast those pathological responses observed after kidney transplantation. Sensitization of T follicular helper cells with the subsequent emergence of detectable donor-specific human leukocyte antigen antibodies, non-human leukocyte antigen antibodies their implication for kidney transplantation and lessons learnt from other transplantation "settings" with special attention to antibody-mediated rejection will be addressed.
Collapse
Affiliation(s)
- Julien Subburayalu
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies (CRTD), Technische Universität Dresden, Dresden, Germany
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
7
|
Filiopoulos V, Vittoraki A, Vallianou K, Bellos I, Markaki P, Liapis G, Marinaki S, Iniotaki A, Boletis IN. The Influence of Antibodies against Angiotensin II Type-1 Receptor on the Outcome of Kidney Transplantation: A Single-Center Retrospective Study. J Clin Med 2023; 12:jcm12093112. [PMID: 37176553 PMCID: PMC10179262 DOI: 10.3390/jcm12093112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Allo- and autoimmune mechanisms are involved in kidney allograft rejection and loss. This study investigates the impact of anti-angiotensin II type-1 receptor antibodies (anti-AT1RAbs) detected alone or in association with HLA donor-specific antibodies (HLA-DSAs) on the outcome of kidney transplantation (KTx). Anti-AT1RAbs and HLA-DSAs were detected in 71 kidney transplant (KT) recipients who developed biopsy-proven acute or chronic active T-cell rejection (TCMR) (n = 51) or antibody-mediated rejection (ABMR) (n = 20), forming the rejection group (RG). The control group (CG) included 71 KTx recipients with comparable characteristics without rejection. All patients had been transplanted with negative T/B flow crossmatch (T/BFCXM). The median follow-up period was 3.7 years. Antibodies were determined pre- and periodically post-KTx by Luminex method for HLA-DSAs and enzyme-linked immunosorbent assay for anti-AT1RAbs. Before KTx, twenty-three (32.4%) patients in the RG, sixteen with TCMR and seven with ABMR, were found anti-AT1Rabs-positive (≥10 U/mL) versus eleven (15.5%) patients in the CG (p = 0.031). Simultaneous detection of preformed anti-AT1RAbs and HLA-DSAs was found in five patients of the RG versus two of the CG (p = 0.355). At the time of transplant biopsy, fifteen (21.1%) patients, four with ABMR and eleven with TCMR, were positive for anti-AT1RAbs. Anti-AT1RAbs and HLA-DSAs were detected simultaneously in 7/15 (46.7%) cases, three with ABMR and four with TCMR. During the follow-up, thirteen (18.3%) patients in the RG, eight with ABMR and five with TCMR, lost their graft compared to one patient (1.4%) in the CG (p = 0.001). Six out of thirteen (46.2%) RG patients who lost the graft were found positive for anti-AT1RAbs pretransplant. Patient survival with functioning graft did not differ significantly between anti-AT1Rabs-positive and negative KT recipients (log-rank p = 0.88). Simultaneous detection of anti-ATR1Abs and HLA-DSAs did not have a significant influence on patient survival with functioning graft (log-rank p = 0.96). Graft function at the end of the follow-up was better, but not significantly, in anti-AT1Rabs-negative patients, with serum creatinine 1.48 [1.20-1.98] mg/dL and eGFR (CKD-EPI) 48.5 [33.5-59.0] mL/min/1.73 m2, compared to anti-AT1Rabs-positive ones who had serum creatinine 1.65 [1.24-2.02] mg/dL (p = 0.394) and eGFR (CKD-EPI) 47.0 [34.8-60.3] mL/min/1.73 m2 (p = 0.966). Anti-AT1RAbs detection pretransplant characterizes KT recipients at increased risk of cellular or antibody-mediated rejection. Furthermore, anti-AT1RAbs, detected alone or simultaneously with HLA-DSAs, appear to be associated with impaired graft function, but their role in graft survival has not been documented in this study. Screening for these antibodies appears to complement pretransplant immunological risk assessment.
Collapse
Affiliation(s)
- Vassilis Filiopoulos
- Clinic of Nephrology and Renal Transplantation, Medical School of Athens, National and Kapodistrian University, Laiko General Hospital, 11527 Athens, Greece
| | - Angeliki Vittoraki
- Immunology Department and National Tissue Typing Center, General Hospital of Athens, 'Georgios Gennimatas', 11527 Athens, Greece
| | - Kalliopi Vallianou
- Clinic of Nephrology and Renal Transplantation, Medical School of Athens, National and Kapodistrian University, Laiko General Hospital, 11527 Athens, Greece
| | - Ioannis Bellos
- Clinic of Nephrology and Renal Transplantation, Medical School of Athens, National and Kapodistrian University, Laiko General Hospital, 11527 Athens, Greece
| | - Pavlina Markaki
- Immunology Department and National Tissue Typing Center, General Hospital of Athens, 'Georgios Gennimatas', 11527 Athens, Greece
| | - George Liapis
- 1st Department of Pathology, Medical School of Athens, National and Kapodistrian University, Laiko General Hospital, 11527 Athens, Greece
| | - Smaragdi Marinaki
- Clinic of Nephrology and Renal Transplantation, Medical School of Athens, National and Kapodistrian University, Laiko General Hospital, 11527 Athens, Greece
| | - Aliki Iniotaki
- Clinic of Nephrology and Renal Transplantation, Medical School of Athens, National and Kapodistrian University, Laiko General Hospital, 11527 Athens, Greece
- Immunology Department and National Tissue Typing Center, General Hospital of Athens, 'Georgios Gennimatas', 11527 Athens, Greece
| | - Ioannis N Boletis
- Clinic of Nephrology and Renal Transplantation, Medical School of Athens, National and Kapodistrian University, Laiko General Hospital, 11527 Athens, Greece
| |
Collapse
|
8
|
Vythoulkas D, Lazana I, Kroupis C, Gavriilaki E, Konstantellos I, Bousiou Z, Chondropoulos S, Griniezaki M, Vardi A, Gkirkas K, Karagiannidou A, Batsis I, Stamouli M, Sakellari I, Tsirigotis P. Endothelial Injury Syndromes after Allogeneic Hematopoietic Stem Cell Transplantation: Angiopetin-2 as a Novel Predictor of the Outcome and the Role of Functional Autoantibodies against Angiotensin II Type 1 and Endothelin A Receptor. Int J Mol Sci 2023; 24:ijms24086960. [PMID: 37108124 PMCID: PMC10138628 DOI: 10.3390/ijms24086960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Transplant-associated thrombotic microangiopathy (TMA) occurs in a significant percentage of patients after allogeneic stem cell transplantation (allo-SCT) and is associated with significant morbidity and mortality. The aim of the present study was to examine the association of serum angiopoetin-2 (Ang2) levels and the presence of antibodies against angiotensin II type 1 (AT1R) and ndothelin A Recreptor (ETAR) with the outcome of patients with TMA and/or graft-versus-host disease (GVHD) after allo-SCT. Analysis of our data showed that elevated serum Ang2 levels at the time of TMA diagnosis are significantly associated with increased non-relapse mortality and decreased overall survival. To our knowledge, this is the first study demonstrating an association between raised Ang2 levels and poor outcomes in patients with TMA. Antibodies against AT1R (AT1R-Abs) and ETAR (ETAR-Abs) were detected in 27% and 23% of the patients, respectively, but there was no association between the presence of autoantibodies and the outcome of patients with TMA. However, a significant finding was the strong positive correlation between the presence of AT1R-Abs with the occurrence of chronic fibrotic GVHD, such as scleroderma and cryptogenic organizing pneumonia, raising the possibility of the contribution of autoantibodies in the pathogenesis of fibrotic GVHD manifestations.
Collapse
Affiliation(s)
- Dionysios Vythoulkas
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ioanna Lazana
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Christos Kroupis
- Clinical Biochemistry and Molecular Diagnostics, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Eleni Gavriilaki
- Hematology and Bone Marrow Transplantation Department, "G. Papanikolaou" General Hospital, 57010 Thessaloniki, Greece
| | - Ioannis Konstantellos
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Zoi Bousiou
- Hematology and Bone Marrow Transplantation Department, "G. Papanikolaou" General Hospital, 57010 Thessaloniki, Greece
| | - Spiros Chondropoulos
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Marianna Griniezaki
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Anna Vardi
- Hematology and Bone Marrow Transplantation Department, "G. Papanikolaou" General Hospital, 57010 Thessaloniki, Greece
| | - Konstantinos Gkirkas
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Aggeliki Karagiannidou
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ioannis Batsis
- Hematology and Bone Marrow Transplantation Department, "G. Papanikolaou" General Hospital, 57010 Thessaloniki, Greece
| | - Maria Stamouli
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ioanna Sakellari
- Hematology and Bone Marrow Transplantation Department, "G. Papanikolaou" General Hospital, 57010 Thessaloniki, Greece
| | - Panagiotis Tsirigotis
- Hematology Division, 2nd Department of Internal Medicine, Propaedeutic, "ATTIKON" University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
9
|
Cabral-Marques O, Moll G, Catar R, Preuß B, Bankamp L, Pecher AC, Henes J, Klein R, Kamalanathan AS, Akbarzadeh R, van Oostveen W, Hohberger B, Endres M, Koolmoes B, Levarht N, Postma R, van Duinen V, van Zonneveld AJ, de Vries-Bouwstra J, Fehres C, Tran F, do Vale FYN, da Silva Souza KB, Filgueiras IS, Schimke LF, Baiocchi GC, de Miranda GC, da Fonseca DLM, Freire PP, Hackel AM, Grasshoff H, Stähle A, Müller A, Dechend R, Yu X, Petersen F, Sotzny F, Sakmar TP, Ochs HD, Schulze-Forster K, Heidecke H, Scheibenbogen C, Shoenfeld Y, Riemekasten G. Autoantibodies targeting G protein-coupled receptors: An evolving history in autoimmunity. Report of the 4th international symposium. Autoimmun Rev 2023; 22:103310. [PMID: 36906052 DOI: 10.1016/j.autrev.2023.103310] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
G protein-coupled receptors (GPCR) are involved in various physiological and pathophysiological processes. Functional autoantibodies targeting GPCRs have been associated with multiple disease manifestations in this context. Here we summarize and discuss the relevant findings and concepts presented in the biennial International Meeting on autoantibodies targeting GPCRs (the 4th Symposium), held in Lübeck, Germany, 15-16 September 2022. The symposium focused on the current knowledge of these autoantibodies' role in various diseases, such as cardiovascular, renal, infectious (COVID-19), and autoimmune diseases (e.g., systemic sclerosis and systemic lupus erythematosus). Beyond their association with disease phenotypes, intense research related to the mechanistic action of these autoantibodies on immune regulation and pathogenesis has been developed, underscoring the role of autoantibodies targeting GPCRs on disease outcomes and etiopathogenesis. The observation repeatedly highlighted that autoantibodies targeting GPCRs could also be present in healthy individuals, suggesting that anti-GPCR autoantibodies play a physiologic role in modeling the course of diseases. Since numerous therapies targeting GPCRs have been developed, including small molecules and monoclonal antibodies designed for treating cancer, infections, metabolic disorders, or inflammatory conditions, anti-GPCR autoantibodies themselves can serve as therapeutic targets to reduce patients' morbidity and mortality, representing a new area for the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Otávio Cabral-Marques
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil; Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil; Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, Brazil.
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany; BIH Center for Regenerative Therapies (BCRT) and Berlin-Brandenburg School for Regenerative Therapies (BSRT), all Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Beate Preuß
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Lukas Bankamp
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Ann-Christin Pecher
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Joerg Henes
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - A S Kamalanathan
- Centre for BioSeparation Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Reza Akbarzadeh
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Wieke van Oostveen
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Endres
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Neurology with Experimental Neurology, Berlin, Germany.; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Stroke Research Berlin, Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), Partner Site Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Bryan Koolmoes
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Nivine Levarht
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Rudmer Postma
- LUMC, Department of Internal Medicine (Nephrology), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden, the Netherlands
| | - Vincent van Duinen
- LUMC, Department of Internal Medicine (Nephrology), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- LUMC, Department of Internal Medicine (Nephrology), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden, the Netherlands
| | - Jeska de Vries-Bouwstra
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Cynthia Fehres
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Florian Tran
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Fernando Yuri Nery do Vale
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gabriela Crispim Baiocchi
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gustavo Cabral de Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Dennyson Leandro Mathias da Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Paula Paccielli Freire
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alexander M Hackel
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Anja Stähle
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Antje Müller
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, A collaboration of Max Delbruck Center for Molecular Medicine and Charité Universitätsmedizin, and HELIOS Clinic, Department of Cardiology and Nephrology, Berlin 13125, Germany
| | - Xinhua Yu
- Priority Area Chronic Lung Diseases, Research Center Borstel (RCB), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Frank Petersen
- Priority Area Chronic Lung Diseases, Research Center Borstel (RCB), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Franziska Sotzny
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Berlin, Germany
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, NY, USA
| | - Hans D Ochs
- University of Washington School of Medicine and Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Carmen Scheibenbogen
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Berlin, Germany
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
10
|
Mawad H, Pinard L, Medani S, Chagnon M, Boucquemont J, Turgeon J, Dieudé M, Hamelin K, Rimbaud AK, Belayachi A, Yang B, Collette S, Sénécal L, Foster BJ, Hébert MJ, Cardinal H. Hypothermic Perfusion Modifies the Association Between Anti-LG3 Antibodies and Delayed Graft Function in Kidney Recipients. Transpl Int 2023; 36:10749. [PMID: 36891519 PMCID: PMC9986256 DOI: 10.3389/ti.2023.10749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 01/26/2023] [Indexed: 02/22/2023]
Abstract
We previously reported associations between autoantibodies to the LG3 fragment of perlecan, anti-LG3, and a higher risk of delayed graft function (DGF) in kidney transplant recipients. Here, we aimed to determine whether some factors that modulate ischemia-reperfusion injury (IRI) can modify this association. We performed a retrospective cohort study in kidney transplant recipients in 2 university-affiliated centers. In 687 patients, we show that high pre-transplant anti-LG3 are associated with DGF when the kidney is transported on ice (odds ratio (OR): 1.75, 95% confidence interval 1.02-3.00), but not when placed on hypothermic perfusion pump (OR: 0.78, 95% CI 0.43-1.37). In patients with DGF, high pre-transplant anti-LG3 are associated with a higher risk of graft failure (subdistribution hazard ratio (SHR): 4.07, 95% CI: 1.80, 9.22), while this was not the case in patients with immediate graft function (SHR: 0.50, 95% CI 0.19, 1.29). High anti-LG3 levels are associated with a higher risk of DGF in kidneys exposed to cold storage, but not when hypothermic pump perfusion is used. High anti-LG3 are also associated with a higher risk of graft failure in patients who experience DGF, a clinical manifestation of severe IRI.
Collapse
Affiliation(s)
- Habib Mawad
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Louis Pinard
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Samar Medani
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Miguel Chagnon
- Department of Mathematics and Statistics, Université de Montréal, Montreal, QC, Canada
| | - Julie Boucquemont
- Montreal Children's Hospital, McGill University, Montreal, QC, Canada
| | - Julie Turgeon
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Mélanie Dieudé
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Héma-Québec, Québec, QC, Canada
| | - Katia Hamelin
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | | | - Ali Belayachi
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Bing Yang
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Suzon Collette
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada.,Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Lynne Sénécal
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada.,Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Bethany J Foster
- Montreal Children's Hospital, McGill University, Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Marie-Josée Hébert
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Héma-Québec, Québec, QC, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Héloïse Cardinal
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Héma-Québec, Québec, QC, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
11
|
Pizzo H, Mirocha J, Choi J, Garrison J, Haas M, Zhang X, Kamil ES, Kim I, Jordan SC, Puliyanda DP. Pre-transplant angiotensin II receptor type I antibodies in pediatric renal transplant recipients: An observational cohort study. Pediatr Transplant 2022; 26:e14400. [PMID: 36168673 DOI: 10.1111/petr.14400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND The role of angiotensin II type 1 receptor antibodies (AT1R-Ab) in pediatric renal transplantation is unclear. Here, we evaluated pre-transplant AT1R-Ab on transplant outcomes in the first 5 years. Secondary analysis compared pre-transplant AT1R-Ab levels by age. METHODS Thirty-six patients, 2-20 years old, were divided into two groups: pre-transplant AT1R-Ab- (<17 U/ml; n = 18) and pre-transplant AT1R-Ab+ (≥17 U/ml; n = 18). eGFR was determined at 6-month, 1-, 2-, and 4-year post-transplant. Allograft biopsies were performed in the setting of strong HLA-DSA (MFI > 10 000), AT1R-Ab ≥17 U/ml, and/or elevated creatinine. RESULTS Mean age in pre-transplant AT1R-Ab- was 13.3 years vs. 11.0 in pre-transplant AT1R-Ab+ (p = 0.16). At 6 months, mean eGFR was 111.3 ml/min/1.73 m2 in pre-transplant AT1R-Ab- vs. 100.2 in pre-transplant AT1R-Ab + at 1 year, 103.6 ml/min/1.73 m2 vs. 100.5; at 2 years, 98.9 ml/min/1.73 m2 vs. and 93.7; at 4 years, 72.6 ml/min/1.73 m2 vs. 80.9. 11/36 patients had acute rejection (6 in pre-transplant AT1R-Ab-, 5 in pre-transplant AT1R-Ab + ). There was no difference in rejection rates. All 6 subjects with de novo HLA-DSA and AT1R-Ab ≥17 U/ml at the time of biopsy experienced rejection. Mean age in those with the AT1R-Ab ≥40 U/ml was 10.0 years vs. 13.2 in those <40 U/ml (p = 0.07). CONCLUSION In our small cohort, pre-transplant AT1R-Ab ≥17 U/ml was not associated with reduced graft function or rejection. The pathogenicity of pre-transplant AT1R-Ab in pediatric kidney transplantation requires further investigation.
Collapse
Affiliation(s)
- Helen Pizzo
- Pediatric Nephrology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - James Mirocha
- Biostatistics Core, Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jua Choi
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jonathan Garrison
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, California, Los Angeles, USA
| | - Xiaohai Zhang
- HLA and Immunogenetics Laboratory, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Elaine S Kamil
- Pediatric Nephrology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Irene Kim
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Stanley C Jordan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dechu P Puliyanda
- Pediatric Nephrology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
12
|
Post-COVID-19 Syndrome: Retinal Microcirculation as a Potential Marker for Chronic Fatigue. Int J Mol Sci 2022; 23:ijms232213683. [PMID: 36430175 PMCID: PMC9690863 DOI: 10.3390/ijms232213683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022] Open
Abstract
Post-COVID-19 syndrome (PCS) is characterized by persisting sequelae after infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). PCS can affect patients with all COVID-19 disease severities. As previous studies have revealed impaired blood flow as a provoking factor triggering PCS, it was the aim of the present study to investigate the potential association between self-reported chronic fatigue and retinal microcirculation in patients with PCS, potentially indicating an objective biomarker. A prospective study was performed, including 201 subjects: 173 patients with PCS and 28 controls. Retinal microcirculation was visualized by OCT angiography (OCT-A) and quantified using the Erlangen-Angio-Tool as macula and peripapillary vessel density (VD). Chronic fatigue (CF) was assessed according to the variables of Bell’s score, age and gender. VDs in the superficial vascular plexus (SVP), intermediate capillary plexus (ICP) and deep capillary plexus (DCP) were analyzed, considering the repetitions (12 times). Seropositivity for autoantibodies targeting G protein-coupled receptors (GPCR-AAbs) was determined by an established cardiomyocyte bioassay. Taking account of the repetitions, a mixed model was performed to detect possible differences in the least square means between the different groups included in the analysis. An age effect in relation to VD was observed between patients and controls (p < 0.0001). Gender analysis showed that women with PCS showed lower VD levels in the SVP compared to male patients (p = 0.0015). The PCS patients showed significantly lower VDs in the ICP as compared to the controls (p = 0.0001 (CI: 0.32; 1)). Moreover, considering PCS patients, the mixed model revealed a significant difference between those with chronic fatigue (CF) and those without CF with respect to VDs in the SVP (p = 0.0033 (CI: −4.5; −0.92)). The model included variables of age, gender and Bell’s score, representing a subjective marker for CF. Consequently, retinal microcirculation might serve as an objective biomarker in subjectively reported chronic fatigue in patients with PCS.
Collapse
|
13
|
Graßhoff H, Fourlakis K, Comdühr S, Riemekasten G. Autoantibodies as Biomarker and Therapeutic Target in Systemic Sclerosis. Biomedicines 2022; 10:2150. [PMID: 36140251 PMCID: PMC9496142 DOI: 10.3390/biomedicines10092150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare connective tissue disorder characterized by immune dysregulation evoking the pathophysiological triad of inflammation, fibrosis and vasculopathy. In SSc, several alterations in the B-cell compartment have been described, leading to polyclonal B-cell hyperreactivity, hypergammaglobulinemia and autoantibody production. Autoreactive B cells and autoantibodies promote and maintain pathologic mechanisms. In addition, autoantibodies in SSc are important biomarkers for predicting clinical phenotype and disease progression. Autoreactive B cells and autoantibodies represent potentially promising targets for therapeutic approaches including B-cell-targeting therapies, as well as strategies for unselective and selective removal of autoantibodies. In this review, we present mechanisms of the innate immune system leading to the generation of autoantibodies, alterations of the B-cell compartment in SSc, autoantibodies as biomarkers and autoantibody-mediated pathologies in SSc as well as potential therapeutic approaches to target these.
Collapse
Affiliation(s)
- Hanna Graßhoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, 23538 Lübeck, Germany
| | | | | | | |
Collapse
|
14
|
Szewczykowski C, Mardin C, Lucio M, Wallukat G, Hoffmanns J, Schröder T, Raith F, Rogge L, Heltmann F, Moritz M, Beitlich L, Schottenhamml J, Herrmann M, Harrer T, Ganslmayer M, Kruse FE, Kräter M, Guck J, Lämmer R, Zenkel M, Gießl A, Hohberger B. Long COVID: Association of Functional Autoantibodies against G-Protein-Coupled Receptors with an Impaired Retinal Microcirculation. Int J Mol Sci 2022; 23:7209. [PMID: 35806214 PMCID: PMC9266742 DOI: 10.3390/ijms23137209] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
Long COVID (LC) describes the clinical phenotype of symptoms after infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnostic and therapeutic options are limited, as the pathomechanism of LC is elusive. As the number of acute SARS-CoV-2 infections was and is large, LC will be a challenge for the healthcare system. Previous studies revealed an impaired blood flow, the formation of microclots, and autoimmune mechanisms as potential factors in this complex interplay. Since functionally active autoantibodies against G-protein-coupled receptors (GPCR-AAbs) were observed in patients after SARS-CoV-2 infection, this study aimed to correlate the appearance of GPCR-AAbs with capillary microcirculation. The seropositivity of GPCR-AAbs was measured by an established cardiomyocyte bioassay in 42 patients with LC and 6 controls. Retinal microcirculation was measured by OCT-angiography and quantified as macula and peripapillary vessel density (VD) by the Erlangen-Angio Tool. A statistical analysis yielded impaired VD in patients with LC compared to the controls, which was accentuated in female persons. A significant decrease in macula and peripapillary VD for AAbs targeting adrenergic β2-receptor, MAS-receptor angiotensin-II-type-1 receptor, and adrenergic α1-receptor were observed. The present study might suggest that a seropositivity of GPCR-AAbs can be linked to an impaired retinal capillary microcirculation, potentially mirroring the systemic microcirculation with consecutive clinical symptoms.
Collapse
Affiliation(s)
- Charlotte Szewczykowski
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Christian Mardin
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Marianna Lucio
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | | | - Jakob Hoffmanns
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Thora Schröder
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Franziska Raith
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Lennart Rogge
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Felix Heltmann
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Michael Moritz
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Lorenz Beitlich
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Julia Schottenhamml
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Martin Herrmann
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.H.); (T.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Thomas Harrer
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.H.); (T.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Marion Ganslmayer
- Department of Internal Medicine 1, Universität of Erlangen-Nürnberg, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Friedrich E. Kruse
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; (M.K.); (J.G.)
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; (M.K.); (J.G.)
| | - Robert Lämmer
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Matthias Zenkel
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Andreas Gießl
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| |
Collapse
|
15
|
Non-HLA Antibodies in Kidney Transplantation: Immunity and Genetic Insights. Biomedicines 2022; 10:biomedicines10071506. [PMID: 35884811 PMCID: PMC9312985 DOI: 10.3390/biomedicines10071506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/27/2022] Open
Abstract
The polymorphic human leukocyte antigen (HLA) system has been considered the main target for alloimmunity, but the non-HLA antibodies and autoimmunity have gained importance in kidney transplantation (KT). Apart from the endothelial injury, secondary self-antigen exposure and the presence of polymorphic alloantigens, respectively, auto- and allo- non-HLA antibodies shared common steps in their development, such as: antigen recognition via indirect pathway by recipient antigen presenting cells, autoreactive T cell activation, autoreactive B cell activation, T helper 17 cell differentiation, loss of self-tolerance and epitope spreading phenomena. Both alloimmunity and autoimmunity play a synergic role in the formation of non-HLA antibodies, and the emergence of transcriptomics and genome-wide evaluation techniques has led to important progress in understanding the mechanistic features. Among them, non-HLA mismatches between donors and recipients provide valuable information regarding the role of genetics in non-HLA antibody immunity and development.
Collapse
|
16
|
Wallukat G, Mattecka S, Wenzel K, Schrödl W, Vogt B, Brunner P, Sheriff A, Kunze R. C-Reactive Protein (CRP) Blocks the Desensitization of Agonistic Stimulated G Protein Coupled Receptors (GPCRs) in Neonatal Rat Cardiomyocytes. J Clin Med 2022; 11:jcm11041058. [PMID: 35207331 PMCID: PMC8878432 DOI: 10.3390/jcm11041058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Recently, C-reactive protein (CRP) was shown to affect intracellular calcium signaling and blood pressure in vitro and in vivo, respectively. The aim of the present study was to further investigate if a direct effect on G-protein coupled receptor (GPCR) signaling by CRP can be observed by using CRP in combination with different GPCR agonists on spontaneously beating cultured neonatal rat cardiomyocytes. All used agonists (isoprenaline, clenbuterol, phenylephrine, angiotensin II and endothelin 1) affected the beat rate of cardiomyocytes significantly and after washing them out and re-stimulation the cells developed a pronounced desensitization of the corresponding receptors. CRP did not affect the basal beating-rate nor the initial increase/decrease in beat-rate triggered by different agonists. However, CRP co-incubated cells did not exhibit desensitization of the respective GPCRs after the stimulation with the different agonists. This lack of desensitization was independent of the GPCR type, but it was dependent on the CRP concentration. Therefore, CRP interferes with the desensitization of GPCRs and has to be considered as a novel regulator of adrenergic, angiotensin-1 and endothelin receptors.
Collapse
Affiliation(s)
- Gerd Wallukat
- Berlin Cures GmbH, BBB Campus, 13125 Berlin, Germany; (G.W.); (K.W.)
| | - Stephan Mattecka
- Pentracor GmbH, 16761 Hennigsdorf, Germany; (S.M.); (B.V.); (P.B.); (A.S.)
| | - Katrin Wenzel
- Berlin Cures GmbH, BBB Campus, 13125 Berlin, Germany; (G.W.); (K.W.)
| | - Wieland Schrödl
- Institute of Bacteriology and Mycology Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany;
| | - Birgit Vogt
- Pentracor GmbH, 16761 Hennigsdorf, Germany; (S.M.); (B.V.); (P.B.); (A.S.)
| | - Patrizia Brunner
- Pentracor GmbH, 16761 Hennigsdorf, Germany; (S.M.); (B.V.); (P.B.); (A.S.)
| | - Ahmed Sheriff
- Pentracor GmbH, 16761 Hennigsdorf, Germany; (S.M.); (B.V.); (P.B.); (A.S.)
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité University Medicine, 12200 Berlin, Germany
| | - Rudolf Kunze
- Pentracor GmbH, 16761 Hennigsdorf, Germany; (S.M.); (B.V.); (P.B.); (A.S.)
- Correspondence:
| |
Collapse
|
17
|
Civieri G, Iop L, Tona F. Antibodies against Angiotensin II Type 1 and Endothelin 1 Type A Receptors in Cardiovascular Pathologies. Int J Mol Sci 2022; 23:ijms23020927. [PMID: 35055116 PMCID: PMC8778295 DOI: 10.3390/ijms23020927] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 12/19/2022] Open
Abstract
Angiotensin II receptor type 1 (AT1R) and endothelin-1 receptor type A (ETAR) are G-protein-coupled receptors (GPCRs) expressed on the surface of a great variety of cells: immune cells, vascular smooth cells, endothelial cells, and fibroblasts express ETAR and AT1R, which are activated by endothelin 1 (ET1) and angiotensin II (AngII), respectively. Certain autoantibodies are specific for these receptors and can regulate their function, thus being known as functional autoantibodies. The function of these antibodies is similar to that of natural ligands, and it involves not only vasoconstriction, but also the secretion of proinflammatory cytokines (such as interleukin-6 (IL6), IL8 and TNF-α), collagen production by fibroblasts, and reactive oxygen species (ROS) release by fibroblasts and neutrophils. The role of autoantibodies against AT1R and ETAR (AT1R-AAs and ETAR-AAs, respectively) is well described in the pathogenesis of many medical conditions (e.g., systemic sclerosis (SSc) and SSc-associated pulmonary hypertension, cystic fibrosis, and allograft dysfunction), but their implications in cardiovascular diseases are still unclear. This review summarizes the current evidence regarding the effects of AT1R-AAs and ETAR-AAs in cardiovascular pathologies, highlighting their roles in heart transplantation and mechanical circulatory support, preeclampsia, and acute coronary syndromes.
Collapse
|
18
|
Hohberger B, Harrer T, Mardin C, Kruse F, Hoffmanns J, Rogge L, Heltmann F, Moritz M, Szewczykowski C, Schottenhamml J, Kräter M, Bergua A, Zenkel M, Gießl A, Schlötzer-Schrehardt U, Lämmer R, Herrmann M, Haberland A, Göttel P, Müller J, Wallukat G. Case Report: Neutralization of Autoantibodies Targeting G-Protein-Coupled Receptors Improves Capillary Impairment and Fatigue Symptoms After COVID-19 Infection. Front Med (Lausanne) 2021; 8:754667. [PMID: 34869451 PMCID: PMC8637609 DOI: 10.3389/fmed.2021.754667] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Clinical features of Coronavirus disease 2019 (COVID-19) are caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Acute infection management is a substantial healthcare issue, and the development of long-Covid syndrome (LCS) is extremely challenging for patients and physicians. It is associated with a variety of characteristics as impaired capillary microcirculation, chronic fatigue syndrome (CFS), proinflammatory cytokines, and functional autoantibodies targeting G-protein-coupled receptors (GPCR-AAbs). Here, we present a case report of successful healing of LCS with BC 007 (Berlin Cures, Berlin, Germany), a DNA aptamer drug with a high affinity to GPCR-AAbs that neutralizes these AAbs. A patient with a documented history of glaucoma, recovered from mild COVID-19, but still suffered from CFS, loss of taste, and impaired capillary microcirculation in the macula and peripapillary region. He was positively tested for various targeting GPCR-AAbs. Within 48 h after a single BC 007 treatment, GPCR-AAbs were functionally inactivated and remained inactive during the observation period of 4 weeks. This observation was accompanied by constant improvement of the fatigue symptoms of the patient, taste, and retinal capillary microcirculation. Therefore, the removal of GPCR-AAb might ameliorate the characteristics of the LCD, such as capillary impairment, loss of taste, and CFS.
Collapse
Affiliation(s)
- Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Harrer
- Max-Planck-Zentrum für Physik und Medizin, Max Planck Institute for the Science of Light, Erlangen, Germany.,Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Mardin
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich Kruse
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Jakob Hoffmanns
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Lennart Rogge
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Felix Heltmann
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Moritz
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Charlotte Szewczykowski
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Schottenhamml
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Kräter
- Max-Planck-Zentrum für Physik und Medizin, Max Planck Institute for the Science of Light, Erlangen, Germany.,Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Antonio Bergua
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Gießl
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Lämmer
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Herrmann
- Max-Planck-Zentrum für Physik und Medizin, Max Planck Institute for the Science of Light, Erlangen, Germany.,Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|
19
|
Catar RA, Wischnewski O, Chen L, Heidecke H, Rutz C, Schülein R, Dragun D, Philippe A, Kusch A. Non-HLA antibodies targeting angiotensin II type 1 receptors and endothelin-1 type A receptors impair endothelial repair via a β2-arrestin link to the mTOR pathway. Kidney Int 2021; 101:498-509. [PMID: 34757123 DOI: 10.1016/j.kint.2021.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023]
Abstract
Functional non-HLA antibodies (antibodies to non-human leukocyte antigens) targeting the G protein-coupled receptors angiotensin II type 1 receptor (AT1R) and endothelin-1 type A receptor (ETAR) are implicated in the pathogenesis of transplant vasculopathy. While ERK signaling (a regulator of cell growth) may represent a general cellular response to agonist stimulation, the molecular link between receptor stimulation and development of vascular obliteration has not been fully established. Here we hypothesize involvement of the versatile adaptor proteins, β-arrestins, and the major regulator of cell growth, PI3K/mTOR signaling, in impaired endothelial repair. To test this, human microvascular endothelial cells were treated with AT1R/ETAR antibodies isolated from patients with kidney transplant vasculopathy. These antibodies activated both mTOR complexes via AT1R and ETAR in a PI3K-dependent and ERK-independent manner. The mTOR inhibitor, rapamycin, completely abolished activation of mTORC1 and mTORC2 after long-term treatment with receptor antibodies. Imaging studies revealed that β2- but not β1-arrestin was recruited to ETAR in response to ET1 and patient antibodies but not with antibodies isolated from healthy individuals. Silencing of β2-arrestin by siRNA transfection significantly reduced ERK1/2 and mTORC2 activation. Non-HLA antibodies impaired endothelial repair by AT1R- and ETAR-induced mTORC2 signaling. Thus, we provide evidence that functional AT1R/ETAR antibodies induce ERK1/2 and mTOR signaling involving β2-arrestin in human microvascular endothelium. Hence, our data may provide a translational rational for mTOR inhibitors in combination with receptor blockers in patients with non-HLA receptor recognizing antibodies.
Collapse
Affiliation(s)
- Rusan Ali Catar
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany.
| | - Oskar Wischnewski
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany
| | - Lei Chen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Department of Nephrology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai City, Guandong Province, People's Republic of China
| | | | - Claudia Rutz
- Leibniz Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin e.V., Berlin, Germany
| | - Ralf Schülein
- Leibniz Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin e.V., Berlin, Germany
| | - Duska Dragun
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Charitéplatz 1, Berlin, Germany
| | - Aurélie Philippe
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Charitéplatz 1, Berlin, Germany
| | - Angelika Kusch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Charitéplatz 1, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.
| |
Collapse
|
20
|
McQuiston A, Emtiazjoo A, Angel P, Machuca T, Christie J, Atkinson C. Set Up for Failure: Pre-Existing Autoantibodies in Lung Transplant. Front Immunol 2021; 12:711102. [PMID: 34456920 PMCID: PMC8385565 DOI: 10.3389/fimmu.2021.711102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/12/2021] [Indexed: 11/17/2022] Open
Abstract
Lung transplant patients have the lowest long-term survival rates compared to other solid organ transplants. The complications after lung transplantation such as primary graft dysfunction (PGD) and ultimately chronic lung allograft dysfunction (CLAD) are the main reasons for this limited survival. In recent years, lung-specific autoantibodies that recognize non-HLA antigens have been hypothesized to contribute to graft injury and have been correlated with PGD, CLAD, and survival. Mounting evidence suggests that autoantibodies can develop during pulmonary disease progression before lung transplant, termed pre-existing autoantibodies, and may participate in allograft injury after transplantation. In this review, we summarize what is known about pulmonary disease autoantibodies, the relationship between pre-existing autoantibodies and lung transplantation, and potential mechanisms through which pre-existing autoantibodies contribute to graft injury and rejection.
Collapse
Affiliation(s)
- Alexander McQuiston
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Amir Emtiazjoo
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Peggi Angel
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Tiago Machuca
- Department of Surgery, University of Florida, Gainesville, FL, United States
| | - Jason Christie
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Carl Atkinson
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
21
|
Freitag H, Szklarski M, Lorenz S, Sotzny F, Bauer S, Philippe A, Kedor C, Grabowski P, Lange T, Riemekasten G, Heidecke H, Scheibenbogen C. Autoantibodies to Vasoregulative G-Protein-Coupled Receptors Correlate with Symptom Severity, Autonomic Dysfunction and Disability in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. J Clin Med 2021; 10:3675. [PMID: 34441971 PMCID: PMC8397061 DOI: 10.3390/jcm10163675] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is an acquired complex disease with patients suffering from the cardinal symptoms of fatigue, post-exertional malaise (PEM), cognitive impairment, pain and autonomous dysfunction. ME/CFS is triggered by an infection in the majority of patients. Initial evidence for a potential role of natural regulatory autoantibodies (AAB) to beta-adrenergic (AdR) and muscarinic acetylcholine receptors (M-AChR) in ME/CFS patients comes from a few studies. METHODS Here, we analyzed the correlations of symptom severity with levels of AAB to vasoregulative AdR, AChR and Endothelin-1 type A and B (ETA/B) and Angiotensin II type 1 (AT1) receptor in a Berlin cohort of ME/CFS patients (n = 116) by ELISA. The severity of disease, symptoms and autonomic dysfunction were assessed by questionnaires. RESULTS We found levels of most AABs significantly correlated with key symptoms of fatigue and muscle pain in patients with infection-triggered onset. The severity of cognitive impairment correlated with AT1-R- and ETA-R-AAB and severity of gastrointestinal symptoms with alpha1/2-AdR-AAB. In contrast, the patients with non-infection-triggered ME/CFS showed fewer and other correlations. CONCLUSION Correlations of specific AAB against G-protein-coupled receptors (GPCR) with symptoms provide evidence for a role of these AAB or respective receptor pathways in disease pathomechanism.
Collapse
Affiliation(s)
- Helma Freitag
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Marvin Szklarski
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Sebastian Lorenz
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Franziska Sotzny
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Sandra Bauer
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Aurélie Philippe
- Department of Nephrology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Claudia Kedor
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Patricia Grabowski
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, 23538 Lübeck, Germany; (T.L.); (G.R.)
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, 23538 Lübeck, Germany; (T.L.); (G.R.)
| | | | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
22
|
Wallukat G, Hohberger B, Wenzel K, Fürst J, Schulze-Rothe S, Wallukat A, Hönicke AS, Müller J. Functional autoantibodies against G-protein coupled receptors in patients with persistent Long-COVID-19 symptoms. J Transl Autoimmun 2021; 4:100100. [PMID: 33880442 PMCID: PMC8049853 DOI: 10.1016/j.jtauto.2021.100100] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/02/2021] [Accepted: 04/10/2021] [Indexed: 02/06/2023] Open
Abstract
Impairment of health after overcoming the acute phase of COVID-19 is being observed more and more frequently. Here different symptoms of neurological and/or cardiological origin have been reported. With symptoms, which are very similar to the ones reported but are not caused by SARS-CoV-2, the occurrence of functionally active autoantibodies (fAABs) targeting G-protein coupled receptors (GPCR-fAABs) has been discussed to be involved. We, therefore investigated, whether GPCR-fAABs are detectable in 31 patients suffering from different Long-COVID-19 symptoms after recovery from the acute phase of the disease. The spectrum of symptoms was mostly of neurological origin (29/31 patients), including post-COVID-19 fatigue, alopecia, attention deficit, tremor and others. Combined neurological and cardiovascular disorders were reported in 17 of the 31 patients. Two recovered COVID-19 patients were free of follow-up symptoms. All 31 former COVID-19 patients had between 2 and 7 different GPCR-fAABs that acted as receptor agonists. Some of those GPCR-fAABs activate their target receptors which cause a positive chronotropic effect in neonatal rat cardiomyocytes, the read-out in the test system for their detection (bioassay for GPCR-fAAB detection). Other GPCR-fAABs, in opposite, cause a negative chronotropic effect on those cells. The positive chronotropic GPCR-fAABs identified in the blood of Long-COVID patients targeted the β2-adrenoceptor (β2-fAAB), the α1-adrenoceptor (α1-fAAB), the angiotensin II AT1-receptor (AT1-fAAB), and the nociceptin-like opioid receptor (NOC-fAAB). The negative chronotropic GPCR-fAABs identified targeted the muscarinic M2-receptor (M2-fAAB), the MAS-receptor (MAS-fAAB), and the ETA-receptor (ETA-fAAB). It was analysed which of the extracellular receptor loops was targeted by the autoantibodies.
Collapse
Key Words
- ACE2, Angiotensin-converting enzyme 2 receptors
- AT1-fAAB, Autoantibody targeting the angiotensin II AT1 receptor
- Autoantibody
- Autoimmunity
- COVID-19
- CRPS, Complex regional pain syndrome
- ETA-fAAB, Autoantibody targeting the endothelin receptor
- Fatigue
- GPCR, G-protein coupled receptors
- Long-COVID
- M2-fAAB, Autoantibody targeting the muscarinic receptor
- MAS-fAAB, Autoantibody targeting the MAS receptor
- NOC-fAAB, Functionally active autoantibody against the nociceptin receptor
- PoTS, Postural orthostatic tachycardia syndrome
- Post-covid-19 symptom
- RAS, Renin angiotensin system
- SARS, Severe acute respiratory syndrome
- fAAB, Functional autoantibody
- α1-fAAB, Autoantibody targeting the alpha1-adrenoceptor
- β2-fAAB, Autoantibody targeting the beta2-adrenoceptor
Collapse
Affiliation(s)
- Gerd Wallukat
- Experimental and Clinical Research Center, Charité Campus Buch, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
- Berlin Cures GmbH, Berlin; Germany
| | - Bettina Hohberger
- Department of Ophthalmology, University of Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Julia Fürst
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | |
Collapse
|
23
|
Sorohan BM, Ismail G, Leca N, Tacu D, Obrișcă B, Constantinescu I, Baston C, Sinescu I. Angiotensin II type 1 receptor antibodies in kidney transplantation: An evidence-based comprehensive review. Transplant Rev (Orlando) 2020; 34:100573. [DOI: 10.1016/j.trre.2020.100573] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
|
24
|
Perez DM. α 1-Adrenergic Receptors in Neurotransmission, Synaptic Plasticity, and Cognition. Front Pharmacol 2020; 11:581098. [PMID: 33117176 PMCID: PMC7553051 DOI: 10.3389/fphar.2020.581098] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
α1-adrenergic receptors are G-Protein Coupled Receptors that are involved in neurotransmission and regulate the sympathetic nervous system through binding and activating the neurotransmitter, norepinephrine, and the neurohormone, epinephrine. There are three α1-adrenergic receptor subtypes (α1A, α1B, α1D) that are known to play various roles in neurotransmission and cognition. They are related to two other adrenergic receptor families that also bind norepinephrine and epinephrine, the β- and α2-, each with three subtypes (β1, β2, β3, α2A, α2B, α2C). Previous studies assessing the roles of α1-adrenergic receptors in neurotransmission and cognition have been inconsistent. This was due to the use of poorly-selective ligands and many of these studies were published before the characterization of the cloned receptor subtypes and the subsequent development of animal models. With the availability of more-selective ligands and the development of animal models, a clearer picture of their role in cognition and neurotransmission can be assessed. In this review, we highlight the significant role that the α1-adrenergic receptor plays in regulating synaptic efficacy, both short and long-term synaptic plasticity, and its regulation of different types of memory. We will also present evidence that the α1-adrenergic receptors, and particularly the α1A-adrenergic receptor subtype, are a potentially good target to treat a wide variety of neurological conditions with diminished cognition.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
25
|
Successful A2 to B Deceased Donor Kidney Transplant after Desensitization for High-Strength Non-HLA Antibody Made Possible by Utilizing a Hepatitis C Positive Donor. Case Rep Transplant 2020; 2020:3591274. [PMID: 32231847 PMCID: PMC7094197 DOI: 10.1155/2020/3591274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/02/2020] [Indexed: 11/28/2022] Open
Abstract
Desensitization using plasma exchange can remove harmful antibodies prior to transplantation and mitigate risks for hyperacute and severe early acute antibody-mediated rejection. Traditionally, the use of plasma exchange requires a living donor so that the timing of treatments relative to transplant can be planned. Non-HLA antibody is increasingly recognized as capable of causing antibody-mediated renal allograft rejection and has been associated with decreased graft longevity. Our patient had high-strength non-HLA antibody deemed prohibitive to transplantation without desensitization, but no living donors. As the patient was eligible to receive an A2 ABO blood group organ and was willing to accept a hepatitis C positive donor kidney, this afforded a high probability of receiving an offer within a short enough time frame to attempt empiric desensitization in anticipation of a deceased donor transplant. Fifteen plasma exchange treatments were performed before the patient received an organ offer, and the patient was successfully transplanted. Hepatitis C infection was treated posttransplant. No episodes of rejection were observed. At one-year posttransplant, the patient maintains good graft function. In this case, willingness to consider nontraditional donor organs enabled us to mimic living donor desensitization using a deceased donor.
Collapse
|
26
|
Philogene MC, Johnson T, Vaught AJ, Zakaria S, Fedarko N. Antibodies against Angiotensin II Type 1 and Endothelin A Receptors: Relevance and pathogenicity. Hum Immunol 2019; 80:561-567. [PMID: 31010696 PMCID: PMC8015780 DOI: 10.1016/j.humimm.2019.04.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 12/25/2022]
Abstract
Antibodies against two G-protein coupled receptors (GPCRs), angiotensin II type 1 receptor (AT1R) and endothelin A receptor (ETAR) are among a growing number of autoantibodies that are found to be associated with allograft dysfunction. AT1R antibodies (AT1Rabs) and ETAR antibodies (ETARabs) have been shown to activate their target receptors and affect signaling pathways. Multiple single center reports have shown an association between presence of these antibodies and acute or chronic rejection and graft loss in kidney, heart, liver, lung and composite tissue transplantations. However, the characteristics of patients that are most likely to develop adverse outcomes, the phenotypes associated with graft damage solely due to these antibodies, and the antibody titer required to cause dysfunction are areas that remain controversial. This review compiles existing knowledge on the effect of antibodies against GPCRs in other diseases in order to bridge the gap in knowledge within transplantation biology. Future areas for research are highlighted and include the need for functional assays and treatment protocols for transplant patients who present with AT1Rabs and ETARabs. Understanding how antibodies that activate GPCRs influence transplantation outcome will have direct clinical implications for preemptive evaluation of transplant candidates as well as the post-transplant care of organ recipients.
Collapse
Affiliation(s)
- Mary Carmelle Philogene
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Tory Johnson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arthur Jason Vaught
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sammy Zakaria
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Neal Fedarko
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
27
|
Han F, Tian P, Xia X, Wang Y, Dou M, Zheng J, Ding X, Xue W, Ding C. Effects of EDC-PEI heparinization on allogeneic vascular antigens and inflammation levels via a rat abdominal aorta transplantation model. Exp Ther Med 2018; 16:5053-5058. [PMID: 30542459 PMCID: PMC6257806 DOI: 10.3892/etm.2018.6879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 09/26/2018] [Indexed: 11/18/2022] Open
Abstract
The objective of this study was to investigate the possibility of increasing the remote patency rate of allogeneic vessel transplantation through studying the effects of N-ethyl-N-(3-dimethylaminopropyl)-carbodiimide and polyethyleneimine (EDC-PEI) heparinization on allogeneic vascular antigens and inflammation levels. Forty rats were randomly divided into the control group, the EDC group, the PEI group and the EDC-PEI group. The rat abdominal aorta was used as the object of study, and the transplanted blood vessels were pretreated with EDC as the water-soluble cross-linking agent and PEI as the heparin-coated carrier. A rat abdominal aorta allogeneic transplantation model was established. Ultrasonic examination was used for observation of patency of proximal and distal anastomosis in each group. The tissue repair after abdominal aorta transplantation in each group was examined by H&E staining. The biomechanics, denaturation temperature and blasting strength of each group were compared. The levels of IL-1, IL-6 and TNF-α in serum of rats were measured by ELISA method, and the expression of MHC-II and α-GAL antigens in blood vessels were detected by immunohistochemistry. There were different degrees of thickening and inflammatory cell aggregation in the abdominal aorta of rats in the control, EDC and PEI groups, but there was no obvious lesion in the EDC-PEI group. Compared with the four groups, the mechanical characteristics of the EDC group decreased significantly, and the stenosis rate of anastomotic stoma in the EDC group was higher than that in the EDC-PEI and PEI groups (P<0.05). The denaturation temperature of the PEI group was lower than that of the EDC and EDC-PEI groups (P<0.05). The mechanical property and vascular bursting strength in the EDC-PEI group were similar to those in the control group. At the same time, it has more significant advantages than the other three groups in removing the vascular antigens MHC-II and α-GAL and reducing the level of inflammatory reaction, thus increasing the remote patency rate of allogeneic vascular transplantation. The inflammatory response and vascular antigenicity after transplantation are effectively reduced via the rat abdominal aorta transplantation model treated with allogeneic EDC-PEI heparinization, which has a higher remote patency rate.
Collapse
Affiliation(s)
- Feng Han
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Puxun Tian
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xinxin Xia
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuxiang Wang
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Meng Dou
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Zheng
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaoming Ding
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wujun Xue
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chenguang Ding
- Department of Kidney Transplantation, Nephropathy Hospital, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China.,Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
28
|
Yamada C, Huang Y, Norman S, Naik A, Moussa O, Samaniego M, Cooling L. Efficacy of therapeutic plasma exchange on angiotensin II type-1 receptor antibodies on two kidney transplant recipients. J Clin Apher 2018; 33:673-677. [DOI: 10.1002/jca.21657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Chisa Yamada
- Division of Transfusion Medicine, Department of Pathology; University of Michigan; Ann Arbor Michigan
| | - Yihung Huang
- Section of Transplant Nephrology, Department of Internal Medicine; University of California, Davis; Davis California
| | - Silas Norman
- Division of Nephrology, Department of Internal Medicine; University of Michigan; Ann Arbor Michigan
| | - Abhijit Naik
- Division of Nephrology, Department of Internal Medicine; University of Michigan; Ann Arbor Michigan
| | - Omar Moussa
- Division of Histocompatibility Laboratory, Department of Pathology; University of Michigan; Ann Arbor Michigan
| | - Milagros Samaniego
- Department of Medicine; Henry Ford Transplant Institute; Detroit Michigan
| | - Laura Cooling
- Division of Transfusion Medicine, Department of Pathology; University of Michigan; Ann Arbor Michigan
| |
Collapse
|
29
|
Cardinal H, Dieudé M, Hébert MJ. Endothelial Dysfunction in Kidney Transplantation. Front Immunol 2018; 9:1130. [PMID: 29875776 PMCID: PMC5974048 DOI: 10.3389/fimmu.2018.01130] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/04/2018] [Indexed: 12/17/2022] Open
Abstract
Kidney transplantation entails a high likelihood of endothelial injury. The endothelium is a target of choice for injury by ischemia-reperfusion, alloantibodies, and autoantibodies. A certain degree of ischemia-reperfusion injury inevitably occurs in the immediate posttransplant setting and can manifest as delayed graft function. Acute rejection episodes, whether T-cell or antibody-mediated, can involve the graft micro- and macrovasculature, leading to endothelial injury and adverse long-term consequences on graft function and survival. In turn, caspase-3 activation in injured and dying endothelial cells favors the release of extracellular vesicles (apoptotic bodies and apoptotic exosome-like vesicles) that further enhance autoantibody production, complement deposition, and microvascular rarefaction. In this review, we present the evidence for endothelial injury, its causes and long-term consequences on graft outcomes in the field of kidney transplantation.
Collapse
Affiliation(s)
- Héloïse Cardinal
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada.,University of Montreal, Montreal, QC, Canada
| | - Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada.,University of Montreal, Montreal, QC, Canada
| |
Collapse
|
30
|
Fichtner A, Süsal C, Schröder C, Höcker B, Rieger S, Waldherr R, Westhoff JH, Sander A, Dragun D, Tönshoff B. Association of angiotensin II type 1 receptor antibodies with graft histology, function and survival in paediatric renal transplant recipients. Nephrol Dial Transplant 2018; 33:1065-1072. [DOI: 10.1093/ndt/gfy008] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/27/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Alexander Fichtner
- Department of Paediatrics I, University Children’s Hospital, Heidelberg, Germany
| | - Caner Süsal
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Claudia Schröder
- Department of Paediatrics I, University Children’s Hospital, Heidelberg, Germany
| | - Britta Höcker
- Department of Paediatrics I, University Children’s Hospital, Heidelberg, Germany
| | - Susanne Rieger
- Department of Paediatrics I, University Children’s Hospital, Heidelberg, Germany
| | - Rüdiger Waldherr
- Department of Nephropathology, Institute of Clinical Pathology, Heidelberg, Germany
| | - Jens H Westhoff
- Department of Paediatrics I, University Children’s Hospital, Heidelberg, Germany
| | - Anja Sander
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Duska Dragun
- Clinic for Nephrology and Critical Care Medicine, Charite-Universitatsmedizin Berlin, corporate member of Freie Universitat Berlin, Humboldt-Universitat zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Burkhard Tönshoff
- Department of Paediatrics I, University Children’s Hospital, Heidelberg, Germany
| |
Collapse
|
31
|
Gunasekaran M, Vachharajani N, Gaut JP, Maw TT, Delos Santos R, Shenoy S, Chapman WC, Wellen J, Mohanakumar T. Development of immune response to tissue-restricted self-antigens in simultaneous kidney-pancreas transplant recipients with acute rejection. Clin Transplant 2017. [PMID: 28639386 DOI: 10.1111/ctr.13009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Simultaneous kidney-pancreas transplantation (SKP Tx) is a treatment for end-stage kidney disease secondary to diabetes mellitus. We investigated the role of immune responses to donor human leukocyte antigens (HLA) and tissue-restricted kidney and pancreas self-antigens (KSAgs and PSAgs, respectively) in SKP Tx recipients (SKP TxRs). Sera collected from 39 SKP TxRs were used to determine de novo Abs specific for KSAgs (collagen-IV, Col-IV; fibronectin, FN) and PSAgs (insulin, islet cells, glutamic acid decarboxylase, and pancreas-associated protein-1) by ELISA. KSAg-specific IFN-γ, IL-17, and IL-10 cytokines were enumerated by ELISpot. Abs to donor HLA classes I and II were determined by Luminex assay. Abs to KSAgs and PSAgs were detectable in recipients with rejection compared with stable recipients (P<.05). Kidney-only rejection recipients had increased Abs against KSAgs compared with stable (P<.05), with no increase in Abs against PSAgs. Pancreas-only rejection recipients showed increased Abs against PSAgs compared to stable (P<.05), with no Abs against KSAgs. SKP TxRs with rejection showed increased frequencies of KSAg-specific IFN-γ and IL-17 with reduction in IL-10-secreting cells. SKP TxRs with rejection developed Abs to KSAgs and PSAgs demonstrated increased frequencies of kidney or pancreas SAg-specific IFN-γ and IL-17-secreting cells with reduced IL-10, suggesting loss of peripheral tolerance to SAgs.
Collapse
Affiliation(s)
| | - Neeta Vachharajani
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph P Gaut
- Department of Anatomic and Molecular Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Thin Thin Maw
- Department of Medicine, Nephrology, University of Southern California, Los Angeles, CA, USA
| | - Rowena Delos Santos
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Surendra Shenoy
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - William C Chapman
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Wellen
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
32
|
Kimball PM, Gupta G, McDougan F. Circulating angiotensin type II receptor: Possible marker for antibody mediated rejection after renal transplantation? Hum Immunol 2017; 78:629-633. [PMID: 28614703 DOI: 10.1016/j.humimm.2017.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/30/2017] [Accepted: 06/08/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Presence of antibody [Ab] against angiotensin receptor [AT1R] indicates heightened risk for antibody mediated rejection [AMR] after transplantation but is insufficient as a marker. We speculated AT1R might be released systemically because of AMR and might be a useful biomarker. METHODS AT1R was measured in blood from 73 Normals and 72 renal patients pre- and post-transplantation. Patients were stratified as AMR-free [Gp1], AMR<1yr [Gp2] and AMR>1yr [Gp3]. RESULTS AT1R was higher [13±26vs.367±537, p<0.01)] and more prevalent [20% vs. 92%, p<0.01] among renal patients than Normals. Pretransplant levels were similar [p=ns] between groups. One-year posttransplant levels approached [p<0.01] normalcy for Gps1+3 but spiked during AMR and remained elevated [155±58, p<0.01] for 50% Gp2 patients. One-year AT1R levels were higher among subsequent graft failures than surviving grafts [171±267vs. 38±50, p<0.01]. CONCLUSIONS Pretransplant AT1R was abnormally elevated: possibly indicating ongoing tissue injury. Pretransplant AT1R didn't predict risk for AMR. However, AT1R spiked during early AMR and sustained elevations were associated with poorer outcomes.
Collapse
Affiliation(s)
- Pamela M Kimball
- Department of Transplant Surgery, VCUHealth Systems, Richmond, VA, United States.
| | - Gaurav Gupta
- Department of Transplant Surgery, VCUHealth Systems, Richmond, VA, United States
| | - Felecia McDougan
- Department of Transplant Surgery, VCUHealth Systems, Richmond, VA, United States
| |
Collapse
|
33
|
Anti-Angiotensin II Type 1 Receptor and Anti-Endothelial Cell Antibodies: A Cross-Sectional Analysis of Pathological Findings in Allograft Biopsies. Transplantation 2017; 101:608-615. [PMID: 27222934 PMCID: PMC5319389 DOI: 10.1097/tp.0000000000001231] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background This is a cross-sectional study designed to evaluate the histologic characteristics of graft injury in the presence of anti-angiotensin II type 1 receptor antibody (AT1R-Ab) and anti-endothelial cell antibody (AECA). Methods Non-HLA antibody testing was included in the posttransplant evaluation for 70 kidney recipients. Biopsies were performed for cause for 47 patients and as protocol for the remaining 23 patients. Biopsy-proven rejection was defined according to the Banff 2009-2013 criteria. AT1R-Ab was measured on an ELISA platform. Patients were divided into 3 groups based on AT1R-Ab levels (>17, 10-17, and <10 U/ml). AECA was evaluated using an endothelial cell crossmatch (ECXM) in patients whose HLA antibody level was insufficient to cause a positive flow cytometric crossmatch. Results AT1R-Ab levels were higher in patients diagnosed with antibody mediated rejection compared to those with no rejection (P = 0.004). Glomerulitis (g) and peritubular capillaritis (ptc) scores were independently correlated with increased AT1R-Ab concentrations in the presence or absence of HLA-DSA (P = 0.007 and 0.03 for g scores; p = 0.005 and 0.03 for ptc scores). Patients with a positive ECXM had higher AT1R-Ab levels compared to those with a negative ECXM (P = 0.005). Microcirculation inflammation (MCI = g + ptc score) was higher in patients with a positive ECXM and with AT1R-Ab >17 U/ml, although this did not reach statistical significance (P = 0.07). Conclusions The data show an association between non-HLA antibodies detected in the ECXM and AT1R ELISA and microvascular injury observed in antibody mediated rejection. The histologic characteristics of kidney graft injury are examined in terms of the presence of anti-angiotensin II type 1 receptor antibody (AT1R-Ab) and anti-endothelial cell antibody (AECA). The presence of AECA or AT1R-Ab correlates with microvascular injury observed in antibody-mediated rejection.
Collapse
|
34
|
Cardiomyopathy - An approach to the autoimmune background. Autoimmun Rev 2017; 16:269-286. [PMID: 28163240 DOI: 10.1016/j.autrev.2017.01.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/20/2016] [Indexed: 12/15/2022]
Abstract
Autoimmunity is increasingly accepted as the origin or amplifier of various diseases. In contrast to classic autoantibodies (AABs), which induce immune responses resulting in the destruction of the affected tissue, an additional class of AABs is directed against G-protein-coupled receptors (GPCRs; GPCR-AABs). GPCR-AABs functionally affect their related GPCRs for activation of receptor mediated signal cascades. Diseases which are characterized by the presence of GPCR-AABs with evidence for disease-specific pathogenic activity could be named "functional autoantibody disease". We briefly summarize here the historical view on autoimmunity in cardiomyopathy, followed by an approach to the mechanistic autoimmunity background. Furthermore, autoantibodies with outstanding importance for cardiomyopathies as a functional autoantibody disease, such as GPCR-AABs, and mainly those directed against the beta1-adrenergic and muscarinic 2 receptor autoantibodies, are introduced. Anti-cardiac myosin and anti-cardiac troponin autoantibodies, as further potential players in autoimmune cardiomyopathy, are additionally taken into account. The basic view on the autoantibodies, their related receptor interactions and pathogenic consequences are presented. Focused specifically on GPCR-AABs, "pros and cons" of assays such as indirect assays (functional changes of cell preparations are monitored after GPCR-AAB receptor binding) and direct assays based on the ELISA technologies (GPCR epitope mimics for GPCR-AAB binding) are critically discussed. Last but not least, treatment strategies for "functional autoantibody disease", such as for GPCR-AAB removal (therapeutic plasma exchange, immunoadsorption) and in vivo GPCR-AAB attack such as intravenous IgG treatment (IVIG), B-cell depletion and GPCR-AAB binding and neutralization, are critically reflected with respect to their patient benefits.
Collapse
|
35
|
Yang B, Dieudé M, Hamelin K, Hénault-Rondeau M, Patey N, Turgeon J, Lan S, Pomerleau L, Quesnel M, Peng J, Tremblay J, Shi Y, Chan JS, Hébert MJ, Cardinal H. Anti-LG3 Antibodies Aggravate Renal Ischemia-Reperfusion Injury and Long-Term Renal Allograft Dysfunction. Am J Transplant 2016; 16:3416-3429. [PMID: 27172087 DOI: 10.1111/ajt.13866] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 05/04/2016] [Accepted: 05/07/2016] [Indexed: 01/25/2023]
Abstract
Pretransplant autoantibodies to LG3 and angiotensin II type 1 receptors (AT1R) are associated with acute rejection in kidney transplant recipients, whereas antivimentin autoantibodies participate in heart transplant rejection. Ischemia-reperfusion injury (IRI) can modify self-antigenic targets. We hypothesized that ischemia-reperfusion creates permissive conditions for autoantibodies to interact with their antigenic targets and leads to enhanced renal damage and dysfunction. In 172 kidney transplant recipients, we found that pretransplant anti-LG3 antibodies were associated with an increased risk of delayed graft function (DGF). Pretransplant anti-LG3 antibodies are inversely associated with graft function at 1 year after transplantation in patients who experienced DGF, independent of rejection. Pretransplant anti-AT1R and antivimentin were not associated with DGF or its functional outcome. In a model of renal IRI in mice, passive transfer of anti-LG3 IgG led to enhanced dysfunction and microvascular injury compared with passive transfer with control IgG. Passive transfer of anti-LG3 antibodies also favored intrarenal microvascular complement activation, microvascular rarefaction and fibrosis after IRI. Our results suggest that anti-LG3 antibodies are novel aggravating factors for renal IRI. These results provide novel insights into the pathways that modulate the severity of renal injury at the time of transplantation and their impact on long-term outcomes.
Collapse
Affiliation(s)
- B Yang
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| | - M Dieudé
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| | - K Hamelin
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| | - M Hénault-Rondeau
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| | - N Patey
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada.,Department of Pathology, CHU Ste-Justine, Université de Montréal, Montreal, QC, Canada
| | - J Turgeon
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| | - S Lan
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| | - L Pomerleau
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - M Quesnel
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - J Peng
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - J Tremblay
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Y Shi
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Université de Montréal, Montreal, QC, Canada
| | - J S Chan
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Université de Montréal, Montreal, QC, Canada
| | - M J Hébert
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| | - H Cardinal
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, T6G 2E1, Canada.,Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
36
|
Cardinal H, Dieudé M, Hébert MJ. The Emerging Importance of Non-HLA Autoantibodies in Kidney Transplant Complications. J Am Soc Nephrol 2016; 28:400-406. [PMID: 27798244 DOI: 10.1681/asn.2016070756] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Antibodies that are specific to organ donor HLA have been involved in the majority of cases of antibody-mediated rejection in solid organ transplant recipients. However, recent data show that production of non-HLA autoantibodies can occur before transplant in the form of natural autoantibodies. In contrast to HLAs, which are constitutively expressed on the cell surface of the allograft endothelium, autoantigens are usually cryptic. Tissue damage associated with ischemia-reperfusion, vascular injury, and/or rejection creates permissive conditions for the expression of cryptic autoantigens, allowing these autoantibodies to bind antigenic targets and further enhance vascular inflammation and renal dysfunction. Antiperlecan/LG3 antibodies and antiangiotensin II type 1 receptor antibodies have been found before transplant in patients with de novo transplants and portend negative long-term outcome in patients with renal transplants. Here, we review mounting evidence suggesting an important role for autoantibodies to cryptic antigens as novel accelerators of kidney dysfunction and acute or chronic allograft rejection.
Collapse
Affiliation(s)
- Héloise Cardinal
- Research Centre, Infection, Inflammation, Immunity and Tissue Injury Axis, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada; and
| | - Mélanie Dieudé
- Research Centre, Infection, Inflammation, Immunity and Tissue Injury Axis, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada; and
| | - Marie-Josée Hébert
- Research Centre, Infection, Inflammation, Immunity and Tissue Injury Axis, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; .,Canadian National Transplant Research Program, Edmonton, Alberta, Canada; and.,Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
37
|
Clinical value of non-HLA antibodies in kidney transplantation: Still an enigma? Transplant Rev (Orlando) 2016; 30:195-202. [DOI: 10.1016/j.trre.2016.06.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/22/2016] [Accepted: 06/01/2016] [Indexed: 12/14/2022]
|
38
|
Bjerre A, Tangeraas T, Heidecke H, Dragun D, Dechend R, Staff AC. Angiotensin II type 1 receptor antibodies in childhood kidney transplantation. Pediatr Transplant 2016; 20:627-32. [PMID: 27251358 DOI: 10.1111/petr.12728] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2016] [Indexed: 12/23/2022]
Abstract
Angiotensin II type 1 receptor antibodies (AT1 RAb) have emerged as non-HLA Ab present in patients with acute AMR and risk of graft loss. Furthermore, AT1 RAb have been shown to increase angiotensin II sensitivity which may play a role in the development of CVD and hypertension. Data on AT1 RAb in stable transplant recipients are lacking. The aim of this study was to analyze the levels of AT1 RAb in a cohort of stable patients after kidney transplantation (tx) in childhood. A cross-sectional study of 30 children (median age 14, range 3-19 yr, median time since tx five yr) and 28 adults who were transplanted in childhood (median age 26, range 20-40 yr, median time since tx 18 yr) transplanted between 1993-2006 and 1983-2002, respectively, was performed. Healthy controls were 51 healthy children (5-8 yr) and 199 healthy donors (median age 56.5 yr, range 42-83 yr). Plasma AT1 RAb were analyzed by immunoassay. Median total AT1 RAb IgG concentration was significantly higher in the pediatric-tx group as compared to the adult-tx group (40.0 and 10.95 U/mL, p < 0.0001). For both groups, the tx group showed higher levels: the pediatric-tx group vs. control group (40.0 vs. 13.3 U/mL, p = 0.0006) and the adult-tx group vs. adult control group (10.95 vs. 6.5 U/mL, p < 0.0001). Age was the strongest indicator of high levels of AT1 RAb IgG (p = 0.0003). AT1 RAb total IgG levels are significantly higher in a stable pediatric-tx cohort as compared to adult-tx patients and healthy controls of comparable age groups. The relevance of our findings in relation to age, time since tx, previous or future rejection, and CVD risk merits future studies.
Collapse
Affiliation(s)
- Anna Bjerre
- Department of Pediatrics, Oslo University Hospital, Oslo, Norway
| | - Trine Tangeraas
- Department of Pediatrics, Oslo University Hospital, Oslo, Norway
| | | | - Duska Dragun
- Department of Nephrology and Intensive Care Medicine, Charité Hospital, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine, and HELIOS Clinic-Berlin, Berlin, Germany
| | - Anne Cathrine Staff
- Department of Obstetrics and Department of Gynecology, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
39
|
Csohány R, Prókai Á, Sziksz E, Balicza-Himer L, Pap D, Kosik A, Sugár D, Vannay Á, Kis-Petik K, Fekete A, Szabó AJ. Sex differences in renin response and changes of capillary diameters after renal ischemia/reperfusion injury. Pediatr Transplant 2016; 20:619-26. [PMID: 27090360 DOI: 10.1111/petr.12712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2016] [Indexed: 01/18/2023]
Abstract
Activation of the RAS has a crucial role in the progression of ischemia/reperfusion-associated CAD. The regulation of RAS differs in the two genders. However, the extent of gender differences and locations of renin production have not been revealed yet. We investigated in vivo the local renin production in the two genders during ischemia/reperfusion injury. In male and female Wistar rats, renal ischemia was induced followed by a reperfusion period of two, eight, 16, 24, or 48 h. We applied flow cytometry to measure renin content and multiphoton imaging to visualize renin granules and changes of peritubular diameters in vivo during ischemia/reperfusion. Renin content decreased in CD in the first eight h of reperfusion; however, after 16 h, its amount increased. In males, the production of renin was more pronounced, and the duration of vasoconstriction was longer with a subsequent phase of vessel hyperdilation compared to females. Renal ischemia/reperfusion injury induces renin response not only in the JGA, but also in the CD segment. Renin production is more explicit in males than in females which, via increased angiotensin II production, might explain the different dynamism of renal vessel regulation between the two genders.
Collapse
Affiliation(s)
- Rózsa Csohány
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Ágnes Prókai
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Erna Sziksz
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | | | - Domonkos Pap
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Anna Kosik
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Dániel Sugár
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Ádám Vannay
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Katalin Kis-Petik
- Institute of Biophysics and Radiational Biology, Semmelweis University, Budapest, Hungary.,MTA-SE Molecular Biophysics Research Group, Budapest, Hungary
| | - Andrea Fekete
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Attila J Szabó
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| |
Collapse
|
40
|
|
41
|
Abstract
The development of post-transplantation antibodies against non-HLA autoantigens is associated with rejection and decreased long-term graft survival. Although our knowledge of non-HLA antibodies is incomplete, compelling experimental and clinical findings demonstrate that antibodies directed against autoantigens such as angiotensin type 1 receptor, perlecan and collagen, contribute to the process of antibody-mediated acute and chronic rejection. The mechanisms that underlie the production of autoantibodies in the setting of organ transplantation is an important area of ongoing investigation. Ischaemia-reperfusion injury, surgical trauma and/or alloimmune responses can result in the release of organ-derived autoantigens (such as soluble antigens, extracellular vesicles or apoptotic bodies) that are presented to B cells in the context of the transplant recipient's antigen presenting cells and stimulate autoantibody production. Type 17 T helper cells orchestrate autoantibody production by supporting the proliferation and maturation of autoreactive B cells within ectopic tertiary lymphoid tissue. Conversely, autoantibody-mediated graft damage can trigger alloimmunity and the development of donor-specific HLA antibodies that can act in synergy to promote allograft rejection. Identification of the immunologic phenotypes of transplant recipients at risk of non-HLA antibody-mediated rejection, and the development of targeted therapies to treat such rejection, are sorely needed to improve both graft and patient survival.
Collapse
|
42
|
Cuevas E, Arreola-Guerra JM, Hernández-Méndez EA, Salcedo I, Castelán N, Uribe-Uribe NO, Vilatobá M, Contreras-Saldívar AG, Sánchez-Cedillo AI, Ramírez JB, de Rungs D, Granados J, Morales-Buenrostro LE, Alberú J. Pretransplant angiotensin II type 1-receptor antibodies are a risk factor for earlier detection of de novo HLA donor-specific antibodies. Nephrol Dial Transplant 2016; 31:1738-45. [PMID: 27220757 DOI: 10.1093/ndt/gfw204] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/05/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Angiotensin II type 1 receptor antibodies (AT1Rabs) have been associated with significantly reduced graft survival. Earlier graft loss has been observed in patients who had pretransplant AT1Rabs and posttransplant donor-specific antibodies (DSA). METHODS The main goal of this retrospective cohort study was to examine the association between AT1Rabs and the time period to detection of de novo human leukocyte antigen (HLA-DSA) posttransplantation in living donor kidney transplant recipients (KTR). The analysis included 141 KTRs. Pretransplant frozen serum samples were tested for AT1Rabs by ELISA and HLA-DSA by SAB (Luminex) at both the pre- and post-KT time points. RESULTS The median AT1Rab level was 9.13 U (interquartile range 5.22-14.33). After a mean follow-up period of 3.55 years, 48 patients were found to harbour de novo HLA-DSAs. The presence of AT1Rabs [hazard ratio (HR) 1.009, 95% confidence interval (CI) 1.002-1.01, P = 0.010], male-to-male transplantation (HR 2.57, 95% CI 1.42-4.67, P = 0.002) and antecedent borderline changes or acute cellular rejection (ACR) (HR 2.47, 95% CI 1.29-4.75, P = 0.006) were significantly associated with de novo DSA detection. A dose-dependent association between AT1Rab levels (<10 U, 10.1-16.9 U, 17-29.9 U and >30 U) and de novo DSA detection was observed (log-rank P = 0.0031). After multivariate analysis of AT1Rab levels (continuous variable), AT1Rabs >30 U, male-to-male transplantation, donor age, higher class I percentage of Panel Reactive Antibody and antecedent borderline changes or ACR remained as independent significant risk factors for the detection of de novo DSAs. CONCLUSIONS The findings suggest that higher levels of pretransplant circulating antibodies against AT1R (>30 U) in kidney graft recipients constitute an independent risk factor for earlier de novo HLA-DSA detection during the posttransplant period.
Collapse
Affiliation(s)
- Eric Cuevas
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - José M Arreola-Guerra
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico Division of Internal Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Erick A Hernández-Méndez
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Isaac Salcedo
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Natalia Castelán
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Norma O Uribe-Uribe
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Mario Vilatobá
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Alan G Contreras-Saldívar
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Aczel I Sánchez-Cedillo
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Julia B Ramírez
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - David de Rungs
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Julio Granados
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Luis E Morales-Buenrostro
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| | - Josefina Alberú
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México, Mexico
| |
Collapse
|
43
|
Dragun D, Catar R, Philippe A. Non-HLA antibodies against endothelial targets bridging allo- and autoimmunity. Kidney Int 2016; 90:280-288. [PMID: 27188505 DOI: 10.1016/j.kint.2016.03.019] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/12/2016] [Accepted: 03/17/2016] [Indexed: 12/17/2022]
Abstract
Detrimental actions of donor-specific antibodies (DSAs) directed against both major histocompatibility antigens (human leukocyte antigen [HLA]) and specific non-HLA antigens expressed on the allograft endothelium are a flourishing research area in kidney transplantation. Newly developed solid-phase assays enabling detection of functional non-HLA antibodies targeting G protein-coupled receptors such as angiotensin type I receptor and endothelin type A receptor were instrumental in providing long-awaited confirmation of their broad clinical relevance. Numerous recent clinical studies implicate angiotensin type I receptor and endothelin type A receptor antibodies as prognostic biomarkers for earlier occurrence and severity of acute and chronic immunologic complications in solid organ transplantation, stem cell transplantation, and systemic autoimmune vascular disease. Angiotensin type 1 receptor and endothelin type A receptor antibodies exert their pathophysiologic effects alone and in synergy with HLA-DSA. Recently identified antiperlecan antibodies are also implicated in accelerated allograft vascular pathology. In parallel, protein array technology platforms enabled recognition of new endothelial surface antigens implicated in endothelial cell activation. Upon target antigen recognition, non-HLA antibodies act as powerful inducers of phenotypic perturbations in endothelial cells via activation of distinct intracellular cell-signaling cascades. Comprehensive diagnostic assessment strategies focusing on both HLA-DSA and non-HLA antibody responses could substantially improve immunologic risk stratification before transplantation, help to better define subphenotypes of antibody-mediated rejection, and lead to timely initiation of targeted therapies. Better understanding of similarities and dissimilarities in HLA-DSA and distinct non-HLA antibody-related mechanisms of endothelial damage should facilitate discovery of common downstream signaling targets and pave the way for the development of endothelium-centered therapeutic strategies to accompany intensified immunosuppression and/or mechanical removal of antibodies.
Collapse
Affiliation(s)
- Duska Dragun
- Clinic for Nephrology and Critical Care Medicine, Campus Virchow-Klinikum and Center for Cardiovascular Research, Medical Faculty of the Charité Berlin, Berlin, Germany; Berlin Institute of Health, Berlin, Germany.
| | - Rusan Catar
- Clinic for Nephrology and Critical Care Medicine, Campus Virchow-Klinikum and Center for Cardiovascular Research, Medical Faculty of the Charité Berlin, Berlin, Germany; Berlin Institute of Health, Berlin, Germany
| | - Aurélie Philippe
- Clinic for Nephrology and Critical Care Medicine, Campus Virchow-Klinikum and Center for Cardiovascular Research, Medical Faculty of the Charité Berlin, Berlin, Germany
| |
Collapse
|
44
|
Bataille A, Payen D, Villiers S, Chazalet JJ, Jacob L. Evolution of Hemodynamic and Functional Human Kidney Graft Dose Response to Dopamine Using an Implantable Doppler Device. EXP CLIN TRANSPLANT 2016; 14:176-83. [PMID: 26767705 DOI: 10.6002/ect.2015.0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES The relation between dopamine infusion and renal hemodynamics and function has not been studied in renal allografts during early recovery. We analyzed the dose response of dopamine infusion on renal blood flow and function in human kidney transplant recipients at reperfusion and during early graft recovery. MATERIALS AND METHODS Phasic and mean renal blood flow was measured by the pulsed Doppler technique using implantable Doppler microprobes in contact with the graft artery. Systemic and renal parameters were recorded on dopamine infusion (0, 3, 5, and 10 μg·kg⁻¹·min⁻¹) immediately after transplant (day 0) in 13 patients and at day 6 in 7/13 patients with early graft recovery. Results are expressed as median and interquartile range between the 25th and 75th percentiles. RESULTS RESULTS At day 0, 3 μg·kg⁻¹·min⁻¹) dopamine did not increase mean renal blood flow over baseline (580 mL/min [219-663 mL/min] vs 542 mL/min [207-686 mL/min]; P = .84). There was an absence of effect with higher dopamine doses, whereas cardiac output, heart rate, and systolic and mean arterial pressure were significantly increased. Urinary sodium excretion, creatinine clearance, and urine output increased dose dependently, with a positive correlation between the increase in urine output and mean arterial pressure (r = 0.48, P < .001). At day 6, 3 μg·kg⁻¹·min⁻¹ dopamine increased mean renal blood flow over baseline (318 mL/min [234-897 mL/min] vs 191 mL/min [173-706 mL/min]; P = .016), with no further increase at higher doses. CONCLUSIONS Immediately after transplant, kidney grafts with ischemic-reperfusion injury are fully dilated and do not respond to dopamine. The specific renal effects observed are due to systemic hemodynamic status. Vascular responsiveness to a "renal dopamine dose" returns on graft recovery.
Collapse
Affiliation(s)
- Aurélien Bataille
- From the Department of Anesthesia and Intensive Care, Groupe Hospitalier Universitaire Saint-Louis-Lariboisière-Fernand-Widal, Assistance Publique-Hôpitaux de Paris, 75010, France
| | | | | | | | | |
Collapse
|
45
|
ABO incompatible renal transplants and decreased likelihood for developing immune responses to HLA and kidney self-antigens. Hum Immunol 2015; 77:76-83. [PMID: 26476207 DOI: 10.1016/j.humimm.2015.10.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/22/2022]
Abstract
Immune responses to HLA and tissue-restricted self-antigens (SAgs) have been proposed to play a role in the pathogenesis of renal allograft (KTx) rejection. However, ABO incompatible (ABOi) KTx recipients (KTxR) following depletion of antibodies (Abs) to blood group antigens had fewer rejections. To determine the mechanisms, pre- and post-transplant sera from ABOi (n=18) and ABO-compatible (ABOc) (n=45) KTxR were analyzed for Abs against HLA class I and II by LABScreen single antigen assay. The development of Abs to SAgs was measured by ELISA. Immunity to Collagen IV (Col-IV) and cytokines induced were measured by ELISPOT. While 8/45 (18%) ABOc KTxR developed new donor specific antibodies to HLA (DSA) following transplantation, 0/18 ABOi KTxR developed DSA. ABOi KTxR failed to develop Abs to kidney SAgs (Col-IV and fibronectin (FN)). In contrast, 7 ABOc KTxR developed Abs to both Col-IV and FN. Col-IV stimulation of lymphocytes from ABOc KTxR demonstrated increased IFNγ, IL-17 and decreased IL-10. In contrast ABOi recipients following stimulation with antigens resulted in more IL10 and reduced IFN-γ and IL17 production. At one year, the GFR in ABOi KTxR were significantly better (p<0.04) than ABOc KTxR. De novo DSA and immune responses to SAgs are reduced or absent in ABOi KTxR which we propose leads to less acute rejection and better long term function following ABOi KTx.
Collapse
|
46
|
Functional autoantibodies in systemic sclerosis. Semin Immunopathol 2015; 37:529-42. [DOI: 10.1007/s00281-015-0513-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/06/2015] [Indexed: 12/29/2022]
|
47
|
Pearl MH, Leuchter RK, Reed EF, Zhang Q, Ettenger RB, Tsai EW. Accelerated rejection, thrombosis, and graft failure with angiotensin II type 1 receptor antibodies. Pediatr Nephrol 2015; 30:1371-4. [PMID: 25956702 PMCID: PMC8514222 DOI: 10.1007/s00467-015-3123-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 11/27/2022]
Abstract
BACKGROUND Angiotensin II type 1 receptor antibodies (AT1R-Abs) have been implicated in renal transplant rejection and failure; however, the mechanism of allograft damage, patterns of clinical presentation, and response to desensitization of AT1R-Abs have not been clearly established. CASE DIAGNOSIS/TREATMENT We present the case of a 7-year-old boy with preformed AT1R-Abs who developed accelerated vascular and cellular rejection and renal allograft thrombosis despite desensitization and treatment with angiotensin receptor blockade. Although an association between AT1R-Abs and microvascular occlusion has been previously described, we are the first to describe an association between AT1R-Abs and renal artery thrombosis, leading to devastating early allograft failure. CONCLUSIONS This case highlights the risk of allograft thrombosis associated with AT1R-Abs and illustrates that previous treatments utilized for AT1R-Abs may not always be effective. Further studies are needed to better characterize the mechanisms of AT1R-Ab pathogenesis and to establish safe levels of AT1R-Abs both pre- and post-transplantation. Given the outcome of this patient and the evidence of pro-coagulatory effects of AT1R-Abs, we suggest that the presence of AT1R-Ab may be a risk factor for thrombosis. The role of treatment with anti-coagulation and novel immunomodulatory agents such as tocilizumab and bortezomib require further investigation.
Collapse
Affiliation(s)
- Meghan H Pearl
- Department of Pediatrics, Division of Nephrology, University of California Los Angeles, David Geffen School of Medicine at UCLA, PO Box 951752, Los Angeles, CA, 90095, USA,
| | | | | | | | | | | |
Collapse
|
48
|
von Rossum A, Laher I, Choy JC. Immune-mediated vascular injury and dysfunction in transplant arteriosclerosis. Front Immunol 2015; 5:684. [PMID: 25628623 PMCID: PMC4290675 DOI: 10.3389/fimmu.2014.00684] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/18/2014] [Indexed: 12/16/2022] Open
Abstract
Solid organ transplantation is the only treatment for end-stage organ failure but this life-saving procedure is limited by immune-mediated rejection of most grafts. Blood vessels within transplanted organs are targeted by the immune system and the resultant vascular damage is a main contributor to acute and chronic graft failure. The vasculature is a unique tissue with specific immunological properties. This review discusses the interactions of the immune system with blood vessels in transplanted organs and how these interactions lead to the development of transplant arteriosclerosis, a leading cause of heart transplant failure.
Collapse
Affiliation(s)
- Anna von Rossum
- Department of Molecular Biology and Biochemistry, Simon Fraser University , Burnaby, BC , Canada
| | - Ismail Laher
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia , Vancouver, BC , Canada
| | - Jonathan C Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University , Burnaby, BC , Canada
| |
Collapse
|
49
|
Becker MO, Kill A, Kutsche M, Guenther J, Rose A, Tabeling C, Witzenrath M, Kühl AA, Heidecke H, Ghofrani HA, Tiede H, Schermuly RT, Nickel N, Hoeper MM, Lukitsch I, Gollasch M, Kuebler WM, Bock S, Burmester GR, Dragun D, Riemekasten G. Vascular Receptor Autoantibodies in Pulmonary Arterial Hypertension Associated with Systemic Sclerosis. Am J Respir Crit Care Med 2014; 190:808-17. [DOI: 10.1164/rccm.201403-0442oc] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
50
|
The patent situation concerning the treatment of diseases associated with autoantibodies directed against G-protein-coupled receptors. Pharm Pat Anal 2014; 2:231-48. [PMID: 24237028 DOI: 10.4155/ppa.12.88] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Agonist-like autoantibodies against receptors of the G-protein-coupled signal cascade have been identified as the pathogenic principle for a variety of diseases, especially those of the heart and vascular system. Consequently, the elimination or neutralization of such autoantibodies is an advised goal for causal therapeutic intervention. This article provides a short, noncomplete overview about remarkable developmental strategies and technical solutions for the therapy of diseases, associated with G-protein-coupled receptor autoantibodies. According to the immunoglobulin nature of the therapeutic target, several strategies are possible, such as the use of the autoantibody epitope sequences as competitors or binding molecules for specific autoantibody-elimination by apheresis. Complete immunoglobulin elimination, as is currently being tested in autoantibody-positive cardiomyopathy patients, would be a nonspecific solution as would the use of immunosuppressant agents. The use of autoantibody-binding molecules on an aptamer basis for neutralization or elimination is a newly developed specific therapeutic option.
Collapse
|