Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Int J Immunopathol Pharmacol ; 35: 20587384211048026, 2021.
Article in English | MEDLINE | ID: mdl-34569339

ABSTRACT

COVID-19 is a highly heterogeneous and complex medical disorder; indeed, severe COVID-19 is probably amongst the most complex of medical conditions known to medical science. While enormous strides have been made in understanding the molecular pathways involved in patients infected with coronaviruses an overarching and comprehensive understanding of the pathogenesis of COVID-19 is lacking. Such an understanding is essential in the formulation of effective prophylactic and treatment strategies. Based on clinical, proteomic, and genomic studies as well as autopsy data severe COVID-19 disease can be considered to be the connection of three basic pathologic processes, namely a pulmonary macrophage activation syndrome with uncontrolled inflammation, a complement-mediated endothelialitis together with a procoagulant state with a thrombotic microangiopathy. In addition, platelet activation with the release of serotonin and the activation and degranulation of mast cells contributes to the hyper-inflammatory state. Auto-antibodies have been demonstrated in a large number of hospitalized patients which adds to the end-organ damage and pro-thrombotic state. This paper provides a clinical overview of the major pathogenetic mechanism leading to severe COVID-19 disease.


Subject(s)
COVID-19/virology , SARS-CoV-2/pathogenicity , COVID-19/blood , COVID-19/immunology , COVID-19/physiopathology , Complement Activation , Complement System Proteins/metabolism , Cytokines/blood , Disease Progression , Host-Pathogen Interactions , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/physiopathology , Inflammation/virology , Inflammation Mediators/blood , Macrophage Activation Syndrome/blood , Macrophage Activation Syndrome/immunology , Macrophage Activation Syndrome/physiopathology , Macrophage Activation Syndrome/virology , Platelet Activation , SARS-CoV-2/immunology , Serotonin/blood , Severity of Illness Index , Thrombotic Microangiopathies/blood , Thrombotic Microangiopathies/immunology , Thrombotic Microangiopathies/physiopathology , Thrombotic Microangiopathies/virology
2.
Br J Haematol ; 193(5): 961-970, 2021 06.
Article in English | MEDLINE | ID: mdl-33954989

ABSTRACT

Descriptions of passenger lymphocyte syndrome (PLS), immune cytopenias and transplant-associated thrombotic microangiopathy (TA-TMA) after intestine-containing transplants remain scarce. We describe our centre's experience of these complications from 2007 to 2019. Ninety-six patients received 103 transplants. PLS occurred in 9 (9%) patients (median 12 days post-transplant); all due to ABO antibodies. There were 31 minor ABO mismatch transplants. No patient required change in immunosuppression. Immune cytopenias (excluding PLS) occurred in six patients at an incidence of 1·7/100 patient years; three immune haemolysis, one immune thrombocytopenia, one acquired Glanzmann's and one immune neutropenia; 50% occurred with other cytopenias. All cases eventually responded to treatment, with a median of four treatments (range 1-8) and 5/6 were treated with rituximab. One patient with immune haemolysis required bortezomib. Complications were common in patients with immune cytopenias; 4/6 with infection needing intravenous antibiotics and 3/6 with venous thromboembolism. In 3/6 cases, a secondary cause for the immune cytopenia was evident. Switching from tacrolimus to ciclosporin was not necessary. There were five cases of transplant-associated thrombotic microangiopathy (TA-TMA; 1·5/100 patient years) requiring calcineurin inhibitor withdrawal; two cases associated with acute rejection. Two cases were managed with plasma exchange, one with plasma infusions and one with eculizumab. Further research in this patient group is required.


Subject(s)
Hemolysis/immunology , Intestines/transplantation , Neutropenia , Organ Transplantation/adverse effects , Thrombasthenia , Thrombotic Microangiopathies , ABO Blood-Group System/immunology , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Bortezomib/administration & dosage , Female , Follow-Up Studies , Humans , Isoantibodies/immunology , Male , Middle Aged , Neutropenia/drug therapy , Neutropenia/etiology , Neutropenia/immunology , Retrospective Studies , Rituximab/administration & dosage , Thrombasthenia/drug therapy , Thrombasthenia/etiology , Thrombasthenia/immunology , Thrombotic Microangiopathies/drug therapy , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/immunology
3.
Medicina (Kaunas) ; 57(3)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799854

ABSTRACT

Renal biopsy is useful to better understand the histological pattern of a lesion (glomerular, tubulointerstitial, and vascular) and the pathogenesis that leads to kidney failure. The potential impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on the kidneys is still undetermined, and a variety of lesions are seen in the kidney tissue of coronavirus disease patients. This review is based on the morphological findings of patients described in case reports and a series of published cases. A search was conducted on MEDLINE and PubMed of case reports and case series of lesions in the presence of non-critical infection by SARS-CoV-2 published until 15/09/2020. We highlight the potential of the virus directly influencing the damage or the innate and adaptive immune response activating cytokine and procoagulant cascades, in addition to the genetic component triggering glomerular diseases, mainly collapsing focal segmental glomerulosclerosis, tubulointerstitial, and even vascular diseases. Kidney lesions caused by SARS-CoV-2 are frequent and have an impact on morbidity and mortality; thus, studies are needed to assess the morphological kidney changes and their mechanisms and may help define their spectrum and immediate or long-term impact.


Subject(s)
Acute Kidney Injury/pathology , COVID-19/pathology , Glomerulonephritis/pathology , Kidney/pathology , Thrombotic Microangiopathies/pathology , Acute Kidney Injury/blood , Acute Kidney Injury/immunology , Adaptive Immunity/immunology , Arteriosclerosis/immunology , Arteriosclerosis/pathology , COVID-19/blood , COVID-19/immunology , Cytokines/immunology , Glomerulonephritis/immunology , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/pathology , Glomerulosclerosis, Focal Segmental/immunology , Glomerulosclerosis, Focal Segmental/pathology , Humans , Immunity, Innate/immunology , Infarction/immunology , Infarction/pathology , Kidney/blood supply , Kidney/immunology , Kidney Cortex Necrosis/immunology , Kidney Cortex Necrosis/pathology , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Nephrosis, Lipoid/immunology , Nephrosis, Lipoid/pathology , Rhabdomyolysis , SARS-CoV-2 , Thrombophilia/blood , Thrombotic Microangiopathies/immunology
4.
Hum Immunol ; 82(5): 371-378, 2021 May.
Article in English | MEDLINE | ID: mdl-33820656

ABSTRACT

Preeclampsia is a multifactorial vascular disease unique to human pregnancy. While genetic and antiangiogenic factors are important contributors to preeclampsia susceptibility, recent studies have shown that dysregulation and/or over-activation of the complement system has an integral role in disease etiology. Furthermore, the role of the coagulation cascade may be underappreciated in the development of the disease. Traditionally, for research purposes, the pool of preeclampsia cases has been divided into non-severe and severe disease depending on the onset and severity of the symptoms. However, of particular interest are a small but important minority of cases that present with symptoms likening to those of hemolysis, elevated liver enzymes and low platelets syndrome, atypical hemolytic uremic syndrome, or thrombotic thrombocytopenic purpura, all thrombotic microangiopathy (TMA) diseases, with the hallmark mechanisms of endothelial dysfunction and aberrant activation of complement and coagulation cascades. We therefore propose a third class, severe TMA-like preeclampsia to be included in the categorization of preeclampsia patients. Identifying these patients would target research, diagnostic differentiation, and novel treatment options to the subclass of patients with life-threatening disease that are most likely to benefit from next-generation drug development.


Subject(s)
Blood Platelets/immunology , Endothelium, Vascular/physiology , Placenta/immunology , Pre-Eclampsia/immunology , Pregnancy/immunology , Thromboinflammation/immunology , Thrombotic Microangiopathies/immunology , Animals , Atypical Hemolytic Uremic Syndrome , Complement Activation , Complement System Proteins/immunology , Complement System Proteins/metabolism , Female , Humans , Models, Immunological
5.
Int J Mol Sci ; 22(8)2021 Apr 18.
Article in English | MEDLINE | ID: mdl-33919576

ABSTRACT

Extracellular vesicles (EV) are microparticles released in biological fluids by different cell types, both in physiological and pathological conditions. Owing to their ability to carry and transfer biomolecules, EV are mediators of cell-to-cell communication and are involved in the pathogenesis of several diseases. The ability of EV to modulate the immune system, the coagulation cascade, the angiogenetic process, and to drive endothelial dysfunction plays a crucial role in the pathophysiology of both autoimmune and renal diseases. Recent studies have demonstrated the involvement of EV in the control of renal homeostasis by acting as intercellular signaling molecules, mediators of inflammation and tissue regeneration. Moreover, circulating EV and urinary EV secreted by renal cells have been investigated as potential early biomarkers of renal injury. In the present review, we discuss the recent findings on the involvement of EV in autoimmunity and in renal intercellular communication. We focused on EV-mediated interaction between the immune system and the kidney in autoimmune diseases displaying common renal damage, such as antiphospholipid syndrome, systemic lupus erythematosus, thrombotic microangiopathy, and vasculitis. Although further studies are needed to extend our knowledge on EV in renal pathology, a deeper investigation of the impact of EV in kidney autoimmune diseases may also provide insight into renal biological processes. Furthermore, EV may represent promising biomarkers of renal diseases with potential future applications as diagnostic and therapeutic tools.


Subject(s)
Antiphospholipid Syndrome/immunology , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Kidney Diseases/immunology , Lupus Erythematosus, Systemic/immunology , Thrombotic Microangiopathies/immunology , Animals , Antiphospholipid Syndrome/metabolism , Humans , Kidney Diseases/metabolism , Lupus Erythematosus, Systemic/metabolism , Thrombotic Microangiopathies/metabolism
8.
Clin Exp Immunol ; 203(1): 96-104, 2021 01.
Article in English | MEDLINE | ID: mdl-32681658

ABSTRACT

Involvement of the alternative complement pathway (AP) in microvascular endothelial cell (MVEC) injury characteristic of a thrombotic microangiopathy (TMA) is well documented. However, the role of the lectin pathway (LP) of complement has not been explored. We examined mannose-binding lectin associated serine protease (MASP2), the effector enzyme of the LP, in thrombotic thrombocytopenic purpura, atypical hemolytic uremic syndrome and post-allogeneic hematopoietic stem cell transplantation (alloHSCT) TMAs. Plasma MASP2 and terminal complement component sC5b-9 levels were assessed by enzyme-linked immunosorbent assay (ELISA). Human MVEC were exposed to patient plasmas, and the effect of the anti-MASP2 human monoclonal antibody narsoplimab on plasma-induced MVEC activation was assessed by caspase 8 activity. MASP2 levels were highly elevated in all TMA patients versus controls. The relatively lower MASP2 levels in alloHSCT patients with TMAs compared to levels in alloHSCT patients who did not develop a TMA, and a significant decrease in variance of MASP2 levels in the former, may reflect MASP2 consumption at sites of disease activity. Plasmas from 14 of the 22 TMA patients tested (64%) induced significant MVEC caspase 8 activation. This was suppressed by clinically relevant levels of narsoplimab (1·2 µg/ml) for all 14 patients, with a mean 65·7% inhibition (36.8-99.4%; P < 0·0001). In conclusion, the LP of complement is activated in TMAs of diverse etiology. Inhibition of MASP2 reduces TMA plasma-mediated MVEC injury in vitro. LP inhibition therefore may be of therapeutic benefit in these disorders.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Endothelial Cells , Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Mannose-Binding Protein-Associated Serine Proteases , Microvessels , Thrombotic Microangiopathies , Adult , Allografts , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Hematologic Neoplasms/blood , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Humans , Male , Mannose-Binding Protein-Associated Serine Proteases/antagonists & inhibitors , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Microvessels/immunology , Microvessels/metabolism , Thrombotic Microangiopathies/blood , Thrombotic Microangiopathies/drug therapy , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/immunology
10.
Nephrol Dial Transplant ; 36(4): 581-586, 2021 03 29.
Article in English | MEDLINE | ID: mdl-31755918

ABSTRACT

Immunoglobulin A nephropathy (IgAN) is considered as mesangiopathy since it initiates in the mesangium; however, other glomerular components are involved and the glomerular capillary wall offers the first contact to circulating macromolecular IgA1. Acute and active forms of IgAN are associated with endocapillary hypercellularity and vascular damage of various degrees, in severe cases with microangiopathy (MA) without or with thrombosis [thrombotic microangiopathy (TMA)]. Vascular damage activates complement and coagulation cascades. A defective complement regulation has recently been detected in active and progressive cases of IgAN. C4d deposits in renal biopsies have been found to be an early risk factor. These observations have raised interest in manifestation of MA and TMA in progressive cases of IgAN. MA-TMA lesions have been found in various percentages (2-53%) of patients with IgAN according to patients' selection and pathology definition of TMA. The association with hypertension (HTN) was so strong that it led to the hypothesis that MA/TMA in IgAN was a mere consequence of severe HTN. Old and new clinical and experimental data indicate that in IgAN the interaction of the glomerular capillary wall with immune reactants and complement uncontrolled activation leading to C4b deposits favours the development of MA-TMA, which plays a role in progression and renal function decline. The central role of complement activation is relevant also for the new therapeutic interventions offered by the pharma.


Subject(s)
Complement Activation/immunology , Complement C4b/immunology , Glomerulonephritis, IGA/pathology , Kidney Glomerulus/pathology , Thrombotic Microangiopathies/pathology , Vascular Diseases/pathology , Glomerulonephritis, IGA/immunology , Humans , Kidney Glomerulus/immunology , Thrombotic Microangiopathies/immunology , Vascular Diseases/immunology
11.
Front Immunol ; 12: 779881, 2021.
Article in English | MEDLINE | ID: mdl-35058924

ABSTRACT

Acute kidney injury (AKI) is a common complication of allogeneic hematopoietic cell transplantation (allo-HCT) and is associated with non-relapse mortality (NRM) and quality of life (QOL). Multiple factors may contribute to AKI during allo-HCT and are often present at the same time making it difficult to determine the cause of AKI in each patient. Nephrotoxic drugs, infections, thrombotic microangiopathy (TMA), and sinusoidal obstruction syndrome (SOS) are well described causes of AKI during allo-HCT. Acute graft-versus-host disease (aGVHD) is a major complication of allo-HCT that mainly targets the intestines, liver, and skin. However, recent studies suggest aGVHD may also attack the kidney and contribute to AKI following allo-HCT. For example, severe aGVHD is associated with AKI, suggesting a link between the two. In addition, animal models have shown donor immune cell infiltration and increased expression of inflammatory cytokines in recipient kidneys after allo-HCT. Therefore, aGVHD may also target the kidney and contribute to AKI following allo-HCT. Herein, we describe the etiology, diagnosis, risk factors, pathophysiology, prevention, and treatment of renal injury after allo-HCT. In addition, we highlight emerging evidence that aGVHD may contribute to the development of AKI after allo-HCT.


Subject(s)
Acute Kidney Injury , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation/adverse effects , Thrombotic Microangiopathies , Tissue Donors , Transplantation Conditioning , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Acute Kidney Injury/therapy , Cytokines/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Humans , Quality of Life , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/immunology
12.
Front Immunol ; 11: 610696, 2020.
Article in English | MEDLINE | ID: mdl-33343584

ABSTRACT

Both neutrophil extracellular traps (NETs) and von Willebrand factor (VWF) are essential for thrombosis and inflammation. During these processes, a complex series of events, including endothelial activation, NET formation, VWF secretion, and blood cell adhesion, aggregation and activation, occurs in an ordered manner in the vasculature. The adhesive activity of VWF multimers is regulated by a specific metalloprotease ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motifs, member 13). Increasing evidence indicates that the interaction between NETs and VWF contributes to arterial and venous thrombosis as well as inflammation. Furthermore, contents released from activated neutrophils or NETs induce the reduction of ADAMTS13 activity, which may occur in both thrombotic microangiopathies (TMAs) and acute ischemic stroke (AIS). Recently, NET is considered as a driver of endothelial damage and immunothrombosis in COVID-19. In addition, the levels of VWF and ADAMTS13 can predict the mortality of COVID-19. In this review, we summarize the biological characteristics and interactions of NETs, VWF, and ADAMTS13, and discuss their roles in TMAs, AIS, and COVID-19. Targeting the NET-VWF axis may be a novel therapeutic strategy for inflammation-associated TMAs, AIS, and COVID-19.


Subject(s)
ADAMTS13 Protein/immunology , COVID-19/immunology , Extracellular Traps/immunology , SARS-CoV-2/immunology , Thrombosis/immunology , von Willebrand Factor/immunology , Acute Disease , Brain Ischemia/immunology , Brain Ischemia/pathology , Brain Ischemia/virology , COVID-19/pathology , Humans , Stroke/immunology , Stroke/pathology , Stroke/virology , Thrombosis/pathology , Thrombosis/virology , Thrombotic Microangiopathies/immunology , Thrombotic Microangiopathies/pathology , Thrombotic Microangiopathies/virology
13.
Am J Case Rep ; 21: e927418, 2020 Sep 12.
Article in English | MEDLINE | ID: mdl-32917848

ABSTRACT

BACKGROUND Patients receiving immunosuppressive therapies might be more susceptible to COVID-19. Conversely, an exaggerated inflammatory response to the SARS-CoV-2 infection might be blunted by certain forms of immunosuppression, which could be protective. Indeed, there are data from animal models demonstrating that complement may be a part of the pathophysiology of coronavirus infections. There is also evidence from an autopsy series demonstrating complement deposition in the lungs of patients with COVID-19. This raises the question of whether patients on anti-complement therapy could be protected from COVID-19. CASE REPORT Case 1 is a 39-year-old woman with an approximately 20-year history of paroxysmal nocturnal hemoglobinuria (PNH), who had recently been switched from treatment with eculizumab to ravulizumab prior to SARS-CoV-2 infection. Case 2 is a 54-year-old woman with a cadaveric renal transplant for lupus nephritis, complicated by thrombotic microangiopathy, who was maintained on eculizumab, which she started several months before she developed the SARS-CoV-2 infection. Case 3 is a 60-year-old woman with a 14-year history of PNH, who had been treated with eculizumab since 2012, and was diagnosed with COVID-19 at the time of her scheduled infusion. All 3 patients had a relatively mild course of COVID-19. CONCLUSIONS We see no evidence of increased susceptibility to SARS-CoV-2 in these patients on anti-complement therapy, which might actually have accounted for the mild course of infection. The effect of anti-complement therapy on COVID-19 disease needs to be determined in clinical trials.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement C5/metabolism , Coronavirus Infections/complications , Hemoglobinuria, Paroxysmal/drug therapy , Pneumonia, Viral/complications , Thrombotic Microangiopathies/drug therapy , Adult , Betacoronavirus , COVID-19 , Complement C5/drug effects , Complement C5/immunology , Complement Inactivating Agents/therapeutic use , Coronavirus Infections/epidemiology , Female , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/immunology , Humans , Middle Aged , Pandemics , Pneumonia, Viral/epidemiology , SARS-CoV-2 , Thrombotic Microangiopathies/complications , Thrombotic Microangiopathies/immunology
14.
Immunobiology ; 225(6): 152001, 2020 11.
Article in English | MEDLINE | ID: mdl-32943233

ABSTRACT

In COVID-19, acute respiratory distress syndrome (ARDS) and thrombotic events are frequent, life-threatening complications. Autopsies commonly show arterial thrombosis and severe endothelial damage. Endothelial damage, which can play an early and central pathogenic role in ARDS and thrombosis, activates the lectin pathway of complement. Mannan-binding lectin-associated serine protease-2 (MASP-2), the lectin pathway's effector enzyme, binds the nucleocapsid protein of severe acute respiratory syndrome-associated coronavirus-2 (SARS-CoV-2), resulting in complement activation and lung injury. Narsoplimab, a fully human immunoglobulin gamma 4 (IgG4) monoclonal antibody against MASP-2, inhibits lectin pathway activation and has anticoagulant effects. In this study, the first time a lectin-pathway inhibitor was used to treat COVID-19, six COVID-19 patients with ARDS requiring continuous positive airway pressure (CPAP) or intubation received narsoplimab under compassionate use. At baseline and during treatment, circulating endothelial cell (CEC) counts and serum levels of interleukin-6 (IL-6), interleukin-8 (IL-8), C-reactive protein (CRP) and lactate dehydrogenase (LDH) were assessed. Narsoplimab treatment was associated with rapid and sustained reduction of CEC and concurrent reduction of serum IL-6, IL-8, CRP and LDH. Narsoplimab was well tolerated; no adverse drug reactions were reported. Two control groups were used for retrospective comparison, both showing significantly higher mortality than the narsoplimab-treated group. All narsoplimab-treated patients recovered and survived. Narsoplimab may be an effective treatment for COVID-19 by reducing COVID-19-related endothelial cell damage and the resultant inflammation and thrombotic risk.


Subject(s)
Antibodies, Monoclonal/therapeutic use , COVID-19/immunology , Complement Pathway, Mannose-Binding Lectin/drug effects , Endothelium, Vascular/drug effects , SARS-CoV-2/immunology , Thrombotic Microangiopathies/drug therapy , Antibodies, Monoclonal/immunology , C-Reactive Protein/immunology , C-Reactive Protein/metabolism , COVID-19/complications , COVID-19/virology , Complement Pathway, Mannose-Binding Lectin/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Inflammation/complications , Inflammation/immunology , Inflammation/prevention & control , Interleukin-6/blood , Interleukin-6/immunology , Male , Mannose-Binding Protein-Associated Serine Proteases/antagonists & inhibitors , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Retrospective Studies , SARS-CoV-2/physiology , Thrombotic Microangiopathies/complications , Thrombotic Microangiopathies/immunology
15.
Nat Rev Rheumatol ; 16(10): 581-589, 2020 10.
Article in English | MEDLINE | ID: mdl-32733003

ABSTRACT

Reports of widespread thromboses and disseminated intravascular coagulation (DIC) in patients with coronavirus disease 19 (COVID-19) have been rapidly increasing in number. Key features of this disorder include a lack of bleeding risk, only mildly low platelet counts, elevated plasma fibrinogen levels, and detection of both severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and complement components in regions of thrombotic microangiopathy (TMA). This disorder is not typical DIC. Rather, it might be more similar to complement-mediated TMA syndromes, which are well known to rheumatologists who care for patients with severe systemic lupus erythematosus or catastrophic antiphospholipid syndrome. This perspective has critical implications for treatment. Anticoagulation and antiviral agents are standard treatments for DIC but are gravely insufficient for any of the TMA disorders that involve disorders of complement. Mediators of TMA syndromes overlap with those released in cytokine storm, suggesting close connections between ineffective immune responses to SARS-CoV-2, severe pneumonia and life-threatening microangiopathy.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/complications , Pneumonia, Viral/complications , Thrombosis/immunology , Anticoagulants/therapeutic use , Antiviral Agents/therapeutic use , COVID-19 , Complement System Proteins/immunology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokines/immunology , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/immunology , Disseminated Intravascular Coagulation/pathology , Disseminated Intravascular Coagulation/virology , Fibrinogen/analysis , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunosuppressive Agents/therapeutic use , Pandemics , Plasma Exchange/methods , Platelet Count/methods , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Risk Factors , SARS-CoV-2 , Thrombosis/drug therapy , Thrombosis/pathology , Thrombosis/virology , Thrombotic Microangiopathies/drug therapy , Thrombotic Microangiopathies/immunology , Thrombotic Microangiopathies/pathology , Thrombotic Microangiopathies/virology
16.
J Clin Immunol ; 40(5): 699-707, 2020 07.
Article in English | MEDLINE | ID: mdl-32447592

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a syndrome of excessive immune system activation driven mainly by high levels of interferon gamma. The clinical presentation of HLH can have considerable overlap with other inflammatory conditions. We present a cohort of patients with therapy refractory HLH referred to our center who were found to have a simultaneous presentation of complement-mediated thrombotic microangiopathy (TMA). Twenty-three patients had therapy refractory HLH (13 primary, 4 EVB-HLH, 6 HLH without known trigger). Sixteen (69.6%) met high-risk TMA criteria. Renal failure requiring renal replacement therapy, severe hypertension, serositis, and gastrointestinal bleeding were documented only in patients with HLH who had concomitant complement-mediated TMA. Patients with HLH and without TMA required ventilator support mainly due to CNS symptoms, while those with HLH and TMA had respiratory failure predominantly associated with pulmonary hypertension, a known presentation of pulmonary TMA. Ten patients received eculizumab for complement-mediated TMA management while being treated for HLH. All patients who received the complement blocker eculizumab in addition to the interferon gamma blocker emapalumab had complete resolution of their TMA and survived. Our observations suggest co-activation of both interferon and complement pathways as a potential culprit in the evolution of thrombotic microangiopathy in patients with inflammatory disorders like refractory HLH and may offer novel therapeutic approaches for these critically ill patients. TMA should be considered in children with HLH and multi-organ failure, as an early institution of a brief course of complement blocking therapy in addition to HLH-targeted therapy may improve clinical outcomes in these patients.


Subject(s)
Interferon-gamma/metabolism , Lymphohistiocytosis, Hemophagocytic/immunology , Thrombotic Microangiopathies/immunology , Adolescent , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/therapeutic use , Child , Child, Preschool , Cohort Studies , Complement Inactivator Proteins/therapeutic use , Complement System Proteins/metabolism , Female , Humans , Hypertension, Pulmonary , Infant , Interferon-gamma/antagonists & inhibitors , Lymphohistiocytosis, Hemophagocytic/mortality , Male , Respiratory Insufficiency , Thrombotic Microangiopathies/mortality , Young Adult
17.
Pathologe ; 41(3): 238-247, 2020 May.
Article in German | MEDLINE | ID: mdl-32240352

ABSTRACT

Increasing interest in the role of the complement system in systemic and renal disease is based on new pathophysiological and therapeutic insights of the recent past and particularly in genetic analyses in children with atypical hemolytic uremic syndrome (aHUS). aHUS is the prototypical systemic disease associated with excessive activation of the alternative complement pathway and manifests in the kidney, but also in other organs as thrombotic microangiopathy (TMA). Pathomechanisms discovered to induce the overactivation of the alternative complement pathway in aHUS led to the first successful therapeutic application of a C5b9 inhibitor. This suppression of the terminal complement cascade succeeded in inhibiting local tissue damage. Thereafter, thanks to advanced modern technologies, further systemic and renal diseases associated with mutations or auto-antibodies targeting the complement pathway were identified. Hereby, disease onset is frequently associated with an additional trigger, e.g. infection or hormonal alterations/imbalances, against the background of a pre-existing predisposition of the patient.Due to the growing understanding of the regulation, and thus the possibility of therapeutic modulation of the different complement pathways, and due to the increasing availability of a variety of drugs inhibiting the complement system, interest in complement-mediated systemic and renal disease has been steadily increasing, making it a "hot-topic" in medicine in recent years.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement Activation , Complement System Proteins/immunology , Kidney Diseases/immunology , Child , Humans , Thrombotic Microangiopathies/immunology
18.
Front Immunol ; 11: 604759, 2020.
Article in English | MEDLINE | ID: mdl-33643292

ABSTRACT

Objective: To first describe and estimate the potential pathogenic role of Ig4 autoantibodies in complement-mediated thrombotic microangiopathy (TMA) in a patient with IgG4-related disease (IgG4-RD). Methods: This study is a case report presenting a retrospective review of the patient's medical chart. Plasma complement C3 and C4 levels, immunoglobulin isotypes and subclasses were determined by nephelometry, the complement pathways' activity (CH50, AP50, MBL) using WIESLAB® Complement System assays. Human complement factor H levels, anti-complement factor H auto-antibodies were analyzed by ELISA, using HRP-labeled secondary antibodies specific for human IgG, IgG4, and IgA, respectively. Genetic analyses were performed by exome sequencing of 14 gens implicated in complement disorders, as well as multiplex ligation-dependent probe amplification looking specifically for CFH, CFHR1-2-3, and 5. Results: Our brief report presents the first case of IgG4-RD with complement-mediated TMA originating from both pathogenic CFHR 1 and CFHR 4 genes deletions, and inhibitory anti-complement factor H autoantibodies of the IgG4 subclass. Remission was achieved with plasmaphereses, corticosteroids, and cyclophosphamide. Following remission, the patient was diagnosed with lymphocytic meningitis and SARS-CoV-2 pneumonia with an uneventful recovery. Conclusion: IgG4-RD can be associated with pathogenic IgG4 autoantibodies. Genetic predisposition such as CFHR1 and CFHR4 gene deletions enhance the susceptibility to the formation of inhibitory anti-Factor H IgG4 antibodies.


Subject(s)
Apolipoproteins/genetics , Atypical Hemolytic Uremic Syndrome/genetics , Autoantibodies/immunology , Complement C3b Inactivator Proteins/genetics , Complement Factor H/immunology , Immunoglobulin G4-Related Disease/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Atypical Hemolytic Uremic Syndrome/pathology , Female , Gene Deletion , Genetic Predisposition to Disease/genetics , Humans , Immunoglobulin G/immunology , Immunoglobulin G4-Related Disease/immunology , Immunoglobulin G4-Related Disease/pathology , Middle Aged , Thrombotic Microangiopathies/immunology , Thrombotic Microangiopathies/pathology
19.
Nephrology (Carlton) ; 25(2): 125-134, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31264312

ABSTRACT

AIM: Renal thrombotic microangiopathy (TMA) is a common pathological manifestation of Castleman's disease (CD)-associated renal lesions. Increased level of plasma vascular endothelial growth factor (VEGF) has been shown in single-case reports. We aimed to investigate the dysregulation of VEGF in the pathogenesis of CD-associated TMA-like lesions (CD-TMA) in a larger cohort. METHODS: Nineteen patients with clinico-pathologically diagnosed CD with renal involvement were enrolled. Ten patients with pregnancy TMA or TMA of unknown reasons were enrolled as TMA control group. The plasma levels of VEGF, soluble Flt-1 and interleukin-6 (IL-6) were detected using enzyme-linked immunosorbent assay kits. The expression of VEGF in the kidney biopsied tissue sections and the lymph node specimens were detected by immunostaining. RESULTS: The plasma levels of VEGF and IL-6 levels were the highest in CD-TMA group compared to TMA control group and healthy controls. The levels of plasma VEGF was positively correlated with that of IL-6, and increased expression of VEGF and IL-6 was also observed in the lymph nodes from CD-TMA patients. However, the expression of VEGF in the glomerular podocytes was significantly decreased in CD-TMA group as well as in the TMA control. CONCLUSION: Our findings suggest that renal VEGF expression might be important in the pathogenetic mechanism of CD-associated TMA-like lesions.


Subject(s)
Castleman Disease , Interleukin-6 , Kidney , Lymph Nodes , Podocytes/immunology , Thrombotic Microangiopathies , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Adult , Biopsy/methods , Castleman Disease/immunology , Castleman Disease/pathology , Female , Humans , Immunohistochemistry , Interleukin-6/analysis , Interleukin-6/blood , Kidney/immunology , Kidney/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Middle Aged , Reproducibility of Results , Thrombotic Microangiopathies/immunology , Thrombotic Microangiopathies/pathology , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-1/analysis , Vascular Endothelial Growth Factor Receptor-1/blood
20.
Exp Clin Transplant ; 18(3): 410-413, 2020 06.
Article in English | MEDLINE | ID: mdl-31615379

ABSTRACT

Lung transplant recipients are at risk of developing many kinds of lung infection, such as community-acquired, nosocomial, opportunistic, and endemic. Here, we report a young lung transplant recipient who developed blastomycosis, which had most likely occurred following eculizumab treatment for atypical hemolytic uremia syndrome. We hypothesize that the agent interfering with C5 would influence the immune response against Blastomyces species. Although eculizumab has opened a new era for treatment of atypical hemolytic uremia syndrome and has led to the understanding that complementmediated pathology is needed, the risk of potentially fatal infections by blocking the complement pathway has not been fully elucidated. Careful follow-up and frequent tests to look for infections are needed after using this monoclonal antibody.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Atypical Hemolytic Uremic Syndrome/drug therapy , Blastomycosis/microbiology , Complement Inactivating Agents/adverse effects , Lung Transplantation/adverse effects , Opportunistic Infections/microbiology , Pulmonary Fibrosis/surgery , Thrombotic Microangiopathies/drug therapy , Antifungal Agents/therapeutic use , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/immunology , Blastomycosis/diagnosis , Blastomycosis/drug therapy , Blastomycosis/immunology , Disease Progression , Fatal Outcome , Humans , Immunocompromised Host , Male , Middle Aged , Opportunistic Infections/diagnosis , Opportunistic Infections/drug therapy , Opportunistic Infections/immunology , Thrombotic Microangiopathies/diagnosis , Thrombotic Microangiopathies/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...