Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Vis Exp ; (191)2023 01 20.
Article in English | MEDLINE | ID: covidwho-2236937

ABSTRACT

An organoid is defined as an engineered multicellular in vitro tissue that mimics its corresponding in vivo organ such that it can be used to study defined aspects of that organ in a tissue culture dish. The breadth and application of human pluripotent stem cell (hPSC)-derived organoid research have advanced significantly to include the brain, retina, tear duct, heart, lung, intestine, pancreas, kidney, and blood vessels, among several other tissues. The development of methods for the generation of human microvessels, specifically, has opened the way for modeling human blood vessel development and disease in vitro and for the testing and analysis of new drugs or tissue tropism in virus infections, including SARS-CoV-2. Complex and lengthy protocols lacking visual guidance hamper the reproducibility of many stem cell-derived organoids. Additionally, the inherent stochasticity of organoid formation processes and self-organization necessitates the generation of optical protocols to advance the understanding of cell fate acquisition and programming. Here, a visually guided protocol is presented for the generation of 3D human blood vessel organoids (BVOs) engineered from hPSCs. Presenting a continuous basement membrane, vascular endothelial cells, and organized articulation with mural cells, BVOs exhibit the functional, morphological, and molecular features of human microvasculature. BVO formation is initiated through aggregate formation, followed by mesoderm and vascular induction. Vascular maturation and network formation are initiated and supported by embedding aggregates in a 3D collagen and solubilized basement membrane matrix. Human vessel networks form within 2-3 weeks and can be further grown in scalable culture systems. Importantly, BVOs transplanted into immunocompromised mice anastomose with the endogenous mouse circulation and specify into functional arteries, veins, and arterioles. The present visually guided protocol will advance human organoid research, particularly in relation to blood vessels in normal development, tissue vascularization, and disease.


Subject(s)
COVID-19 , Pluripotent Stem Cells , Humans , Animals , Mice , Endothelial Cells/physiology , Reproducibility of Results , SARS-CoV-2 , Organoids , Cell Differentiation/physiology
2.
Cells ; 11(10)2022 05 20.
Article in English | MEDLINE | ID: covidwho-1869480

ABSTRACT

Successful neuroprotection is only possible with contemporary microvascular protection. The prevention of disease-induced vascular modifications that accelerate brain damage remains largely elusive. An improved understanding of pericyte (PC) signalling could provide important insight into the function of the neurovascular unit (NVU), and into the injury-provoked responses that modify cell-cell interactions and crosstalk. Due to sharing the same basement membrane with endothelial cells, PCs have a crucial role in the control of endothelial, astrocyte, and oligodendrocyte precursor functions and hence blood-brain barrier stability. Both cerebrovascular and neurodegenerative diseases impair oxygen delivery and functionally impair the NVU. In this review, the role of PCs in central nervous system health and disease is discussed, considering their origin, multipotency, functions and also dysfunction, focusing on new possible avenues to modulate neuroprotection. Dysfunctional PC signalling could also be considered as a potential biomarker of NVU pathology, allowing us to individualize therapeutic interventions, monitor responses, or predict outcomes.


Subject(s)
Endothelial Cells , Pericytes , Astrocytes , Blood-Brain Barrier/pathology , Cell Communication , Endothelial Cells/physiology , Pericytes/pathology
3.
Oxid Med Cell Longev ; 2022: 1630918, 2022.
Article in English | MEDLINE | ID: covidwho-1714452

ABSTRACT

BACKGROUND: The impairment of microcirculation is associated with the unfavorable outcome for extracorporeal membrane oxygenation (ECMO) patients. Studies revealed that pulsatile modification improves hemodynamics and attenuates inflammation during ECMO support. However, whether flow pattern impacts microcirculation and endothelial integrity is rarely documented. The objective of this work was to explore how pulsatility affects microcirculation during ECMO. METHODS: Canine animal models with cardiac arrest were supported by ECMO, with the i-Cor system used to generate nonpulsatile or pulsatile flow. The sublingual microcirculation parameters were examined using the CytoCam microscope system. The expression of hsa_circ_0007367, a circular RNA, was measured during ECMO support. In vitro validation was performed in pulmonary vascular endothelial cells (PMVECs) exposed to pulsatile or nonpulsatile flow, and the expressions of hsa_circ_0007367, endothelial tight junction markers, endothelial adhesive molecules, endothelial nitric oxide synthases (eNOS), and NF-κB signaling activity were analyzed. RESULTS: The pulsatile modification of ECMO enhanced microcirculatory perfusion, attenuated pulmonary inflammation, and stabilized endothelial integrity in animal models; meanwhile, the expression of hsa_circ_0007367 was significantly upregulated both in animals and PMVECs exposed to pulsatile flow. In particular, upregulation of hsa_circ_0007367 stabilized the expressions of endothelial tight junction markers zonula occludens- (ZO-) 1 and occludin, followed by modulating the endothelial nitric oxide synthases (eNOS) activity and inhibiting the NF-κB signaling pathway. CONCLUSION: The modification of pulsatility contributes to microcirculatory perfusion and endothelial integrity during ECMO. The expression of hsa_circ_0007367 plays a pivotal role in this protective mechanism.


Subject(s)
Cell-Free Nucleic Acids/genetics , Endothelial Cells/physiology , Extracorporeal Membrane Oxygenation/methods , Heart Arrest/therapy , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dogs , Endothelial Cells/metabolism , Heart Arrest/genetics , Heart Arrest/pathology , Heart Arrest/physiopathology , Inflammation , Lung/blood supply , Lung/pathology , Microcirculation , Nitric Oxide Synthase Type III/metabolism , Occludin/genetics , Occludin/metabolism , Pulsatile Flow , Rats , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
4.
J Am Soc Nephrol ; 32(2): 479-494, 2021 02.
Article in English | MEDLINE | ID: covidwho-1496663

ABSTRACT

BACKGROUND: Binding of donor-specific antibodies (DSAs) to kidney allograft endothelial cells that does not activate the classic complement cascade can trigger the recruitment of innate immune effectors, including NK cells. Activated NK cells contribute to microvascular inflammation leading to chronic antibody-mediated rejection (AMR). Recipient NK cells can also trigger antibody-independent microvascular inflammation by sensing the absence of self HLA class I molecules ("missing self") on allograft endothelial cells. This translational study investigated whether the condition of missing self amplifies DSA-dependent NK cell activation to worsen chronic AMR. METHODS AND RESULTS: Among 1682 kidney transplant recipients who underwent an allograft biopsy at Lyon University Hospital between 2004 and 2017, 135 fulfilled the diagnostic criteria for AMR and were enrolled in the study. Patients with complement-fixing DSAs identified by a positive C3d binding assay (n=73, 54%) had a higher risk of transplant failure (P=0.002). Among the remaining patients with complement-independent chronic AMR (n=62, 46%), those in whom missing self was identified through donor and recipient genotyping exhibited worse allograft survival (P=0.02). In multivariable analysis, only proteinuria (HR: 7.24; P=0.01) and the presence of missing self (HR: 3.57; P=0.04) were independent predictors for transplant failure following diagnosis of chronic AMR. Cocultures of human NK cells and endothelial cells confirmed that addition of missing self to DSA-induced NK cell activation increased endothelial damage. CONCLUSIONS: The assessment of missing self at the time of diagnosis of chronic AMR identifies patients at higher risk for kidney transplant failure.


Subject(s)
Allografts/pathology , Complement Activation/physiology , Graft Rejection/etiology , Histocompatibility Antigens Class I/blood , Kidney Transplantation/adverse effects , Killer Cells, Natural/physiology , Adult , Allografts/immunology , Cell Culture Techniques , Complement C3d/metabolism , Endothelial Cells/physiology , Female , Graft Rejection/blood , Graft Rejection/pathology , Graft Survival , Humans , Killer Cells, Natural/pathology , Male , Middle Aged , Young Adult
5.
Methods Mol Biol ; 2311: 185-193, 2021.
Article in English | MEDLINE | ID: covidwho-1482181

ABSTRACT

Studies of blood-brain barrier (BBB) require developing of a novel and convenient in vitro endothelial cell model. We isolated primary human and rodent brain microvascular endothelial cells and developed methods for culturing, characterization, and high-efficiency transfection of endothelial cells. Here, we describe the improved methods to obtain in vitro human and rodent BBB models to study expression of endogenous and exogenous genes of interest.


Subject(s)
Blood-Brain Barrier/physiology , Brain/blood supply , Cell Separation , Endothelial Cells/physiology , Microvessels/cytology , Transfection , Animals , Blood-Brain Barrier/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Fetus , Gestational Age , Humans , Mice , Rats
6.
Front Immunol ; 12: 653110, 2021.
Article in English | MEDLINE | ID: covidwho-1305643

ABSTRACT

To characterize transcriptomic changes in endothelial cells (ECs) infected by coronaviruses, and stimulated by DAMPs, the expressions of 1311 innate immune regulatomic genes (IGs) were examined in 28 EC microarray datasets with 7 monocyte datasets as controls. We made the following findings: The majority of IGs are upregulated in the first 12 hours post-infection (PI), and maintained until 48 hours PI in human microvascular EC infected by middle east respiratory syndrome-coronavirus (MERS-CoV) (an EC model for COVID-19). The expressions of IGs are modulated in 21 human EC transcriptomic datasets by various PAMPs/DAMPs, including LPS, LPC, shear stress, hyperlipidemia and oxLDL. Upregulation of many IGs such as nucleic acid sensors are shared between ECs infected by MERS-CoV and those stimulated by PAMPs and DAMPs. Human heart EC and mouse aortic EC express all four types of coronavirus receptors such as ANPEP, CEACAM1, ACE2, DPP4 and virus entry facilitator TMPRSS2 (heart EC); most of coronavirus replication-transcription protein complexes are expressed in HMEC, which contribute to viremia, thromboembolism, and cardiovascular comorbidities of COVID-19. ECs have novel trained immunity (TI), in which subsequent inflammation is enhanced. Upregulated proinflammatory cytokines such as TNFα, IL6, CSF1 and CSF3 and TI marker IL-32 as well as TI metabolic enzymes and epigenetic enzymes indicate TI function in HMEC infected by MERS-CoV, which may drive cytokine storms. Upregulated CSF1 and CSF3 demonstrate a novel function of ECs in promoting myelopoiesis. Mechanistically, the ER stress and ROS, together with decreased mitochondrial OXPHOS complexes, facilitate a proinflammatory response and TI. Additionally, an increase of the regulators of mitotic catastrophe cell death, apoptosis, ferroptosis, inflammasomes-driven pyroptosis in ECs infected with MERS-CoV and the upregulation of pro-thrombogenic factors increase thromboembolism potential. Finally, NRF2-suppressed ROS regulate innate immune responses, TI, thrombosis, EC inflammation and death. These transcriptomic results provide novel insights on the roles of ECs in coronavirus infections such as COVID-19, cardiovascular diseases (CVD), inflammation, transplantation, autoimmune disease and cancers.


Subject(s)
Coronavirus Infections/immunology , Cytokine Release Syndrome/immunology , Endothelial Cells/physiology , Inflammation/immunology , Middle East Respiratory Syndrome Coronavirus/physiology , NF-E2-Related Factor 2/metabolism , SARS-CoV-2/physiology , Alarmins/immunology , Animals , Datasets as Topic , Endothelial Cells/virology , Gene Expression Profiling , Humans , Immunity, Innate , Immunization , Mice , Myelopoiesis , Oxidative Stress , Thromboembolism
7.
Front Immunol ; 12: 634416, 2021.
Article in English | MEDLINE | ID: covidwho-1305641

ABSTRACT

BACKGROUND: The coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and has evoked a pandemic that challenges public health-care systems worldwide. Endothelial cell dysfunction plays a key role in pathophysiology, and simple prognosticators may help to optimize allocation of limited resources. Endothelial activation and stress index (EASIX) is a validated predictor of endothelial complications and outcome after allogeneic stem cell transplantation. Aim of this study was to test if EASIX could predict life-threatening complications in patients with COVID-19. METHODS: SARS-CoV-2-positive, hospitalized patients were enrolled onto a prospective non-interventional register study (n=100). Biomarkers were assessed at hospital admission. Primary endpoint was severe course of disease (mechanical ventilation and/or death, V/D). Results were validated in 126 patients treated in two independent institutions. RESULTS: EASIX at admission was a strong predictor of severe course of the disease (odds ratio for a two-fold change 3.4, 95%CI 1.8-6.3, p<0.001), time to V/D (hazard ratio (HR) for a two-fold change 2.0, 95%CI 1.5-2.6, p<0.001) as well as survival (HR for a two-fold change 1.7, 95%CI 1.2-2.5, p=0.006). The effect was retained in multivariable analysis adjusting for age, gender, and comorbidities and could be validated in the independent cohort. At hospital admission EASIX correlated with increased suppressor of tumorigenicity-2, soluble thrombomodulin, angiopoietin-2, CXCL8, CXCL9 and interleukin-18, but not interferon-alpha. CONCLUSION: EASIX is a validated predictor of COVID19 outcome and an easy-to-access tool to segregate patients in need for intensive surveillance.


Subject(s)
COVID-19/diagnosis , Endothelial Cells/physiology , Hematopoietic Stem Cell Transplantation , SARS-CoV-2/physiology , Severity of Illness Index , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , COVID-19/mortality , Female , Hospitalization , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Respiration, Artificial , Survival Analysis , Transplantation, Homologous , Treatment Outcome , Young Adult
8.
J Am Soc Nephrol ; 31(2): 297-307, 2020 02.
Article in English | MEDLINE | ID: covidwho-992922

ABSTRACT

BACKGROUND: Myeloperoxidase-specific ANCA (MPO-ANCA) are implicated in the pathogenesis of vasculitis and GN. Kinins play a major role during acute inflammation by regulating vasodilatation and vascular permeability and by modulating adhesion and migration of leukocytes. Kinin system activation occurs in patients with ANCA vasculitis. Previous studies in animal models of GN and sclerosing kidney diseases have demonstrated protective effects of bradykinin receptor 1 (B1R) blockade via interference with myeloid cell trafficking. METHODS: To investigate the role of B1R in a murine model of MPO-ANCA GN, we evaluated effects of B1R genetic ablation and pharmacologic blockade. We used bone marrow chimeric mice to determine the role of B1R in bone marrow-derived cells (leukocytes) versus nonbone marrow-derived cells. We elucidated mechanisms of B1R effects using in vitro assays for MPO-ANCA-induced neutrophil activation, endothelial adherence, endothelial transmigration, and neutrophil adhesion molecule surface display. RESULTS: B1R deficiency or blockade prevented or markedly reduced ANCA-induced glomerular crescents, necrosis, and leukocyte influx in mice. B1R was not required for in vitro MPO-ANCA-induced neutrophil activation. Leukocyte B1R deficiency, but not endothelial B1R deficiency, decreased glomerular neutrophil infiltration induced by MPO-ANCA in vivo. B1R enhanced ANCA-induced neutrophil endothelial adhesion and transmigration in vitro. ANCA-activated neutrophils exhibited changes in Mac-1 and LFA-1, important regulators of neutrophil endothelial adhesion and transmigration: ANCA-activated neutrophils increased surface expression of Mac-1 and increased shedding of LFA-1, whereas B1R blockade reduced these effects. CONCLUSIONS: The leukocyte B1R plays a critical role in the pathogenesis of MPO-ANCA-induced GN in a mouse model by modulating neutrophil-endothelial interaction. B1R blockade may have potential as a therapy for ANCA GN and vasculitis.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Glomerulonephritis/etiology , Peroxidase/immunology , Receptor, Bradykinin B1/physiology , Animals , Bradykinin B1 Receptor Antagonists/therapeutic use , Cell Adhesion , Disease Models, Animal , Endothelial Cells/physiology , Glomerulonephritis/drug therapy , Mice , Mice, Inbred C57BL , Neutrophils/physiology
9.
Life Sci ; 264: 118617, 2021 Jan 01.
Article in English | MEDLINE | ID: covidwho-880558

ABSTRACT

BACKGROUND: COVID-19-associated acute respiratory distress syndrome (ARDS) is associated with significant morbidity and high levels of mortality. This paper describes the processes involved in the pathophysiology of COVID-19 from the initial infection and subsequent destruction of type II alveolar epithelial cells by SARS-CoV-2 and culminating in the development of ARDS. MAIN BODY: The activation of alveolar cells and alveolar macrophages leads to the release of large quantities of proinflammatory cytokines and chemokines and their translocation into the pulmonary vasculature. The presence of these inflammatory mediators in the vascular compartment leads to the activation of vascular endothelial cells platelets and neutrophils and the subsequent formation of platelet neutrophil complexes. These complexes in concert with activated endothelial cells interact to create a state of immunothrombosis. The consequence of immunothrombosis include hypercoagulation, accelerating inflammation, fibrin deposition, migration of neutrophil extracellular traps (NETs) producing neutrophils into the alveolar apace, activation of the NLRP3 inflammazome, increased alveolar macrophage destruction and massive tissue damage by pyroptosis and necroptosis Therapeutic combinations aimed at ameliorating immunothrombosis and preventing the development of severe COVID-19 are discussed in detail.


Subject(s)
COVID-19/immunology , COVID-19/physiopathology , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/prevention & control , SARS-CoV-2/pathogenicity , Thrombosis/complications , Thrombosis/physiopathology , Alveolar Epithelial Cells/physiology , Blood Platelets/physiology , COVID-19/complications , Cytokines/physiology , Endothelial Cells/physiology , Humans , Macrophages, Alveolar/physiology , Neutrophils/physiology , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Thrombosis/immunology , COVID-19 Drug Treatment
10.
Cardiovasc Drugs Ther ; 35(3): 411-425, 2021 06.
Article in English | MEDLINE | ID: covidwho-756483

ABSTRACT

The recent emergence of the coronavirus disease 19 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in China is now a global health emergency. The transmission of SARS-CoV-2 is mainly via human-to-human contact. This virus is expected to be of zoonotic origin and has a high genome identity to that of bat derived SARS-like coronavirus. Various stringent measures have been implemented to lower person-to-person transmission of COVID-19. Particular observations and attempts have been made to reduce transmission in vulnerable populations, including older adults, children, and healthcare providers. This novel CoV enters the cells through the angiotensin-converting enzyme 2 (ACE2) receptor. There is a higher risk of COVID-19 infection among those with preexisting cardiovascular diseases (CVD), and it has been connected with various direct and indirect complications, including myocarditis, acute myocardial injury, venous thromboembolism, and arrhythmias. This article summarizes the various cardiovascular complications and mechanisms responsible for the same with COVID-19 infection. For the benefit of the scientific community and public, the effect of COVID-19 on major vital organs such as the kidneys, liver, and intestines has been briefly discussed. In this review, we also discuss drugs in different stages of clinical trials and their associated complications, as well as the details of vaccines in various stages of development.


Subject(s)
COVID-19/complications , Cardiovascular Diseases/complications , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/physiology , Antiviral Agents/adverse effects , COVID-19/immunology , Cytokine Release Syndrome/etiology , Endothelial Cells/physiology , Humans , Hypertension/complications , Immunity, Innate , Myocardial Infarction/complications , Sex Characteristics , Thrombophilia/etiology , COVID-19 Drug Treatment
11.
Innate Immun ; 27(2): 109-117, 2021 02.
Article in English | MEDLINE | ID: covidwho-751270

ABSTRACT

The blood-brain barrier (BBB), which protects the CNS from pathogens, is composed of specialized brain microvascular endothelial cells (BMECs) joined by tight junctions and ensheathed by pericytes and astrocyte endfeet. The stability of the BBB structure and function is of great significance for the maintenance of brain homeostasis. When a neurotropic virus invades the CNS via a hematogenous or non-hematogenous route, it may cause structural and functional disorders of the BBB, and also activate the BBB anti-inflammatory or pro-inflammatory innate immune response. This article focuses on the structural and functional changes that occur in the three main components of the BBB (endothelial cells, astrocytes, and pericytes) in response to infection with neurotropic viruses transmitted by hematogenous routes, and also briefly describes the supportive effect of three cells on the BBB under normal physiological conditions. For example, all three types of cells express several PRRs, which can quickly sense the virus and make corresponding immune responses. The pro-inflammatory immune response will exacerbate the destruction of the BBB, while the anti-inflammatory immune response, based on type I IFN, consolidates the stability of the BBB. Exploring the details of the interaction between the host and the pathogen at the BBB during neurotropic virus infection will help to propose new treatments for viral encephalitis. Enhancing the defense function of the BBB, maintaining the integrity of the BBB, and suppressing the pro-inflammatory immune response of the BBB provide more ideas for limiting the neuroinvasion of neurotropic viruses. In the future, these new treatments are expected to cooperate with traditional antiviral methods to improve the therapeutic effect of viral encephalitis.


Subject(s)
Blood-Brain Barrier/physiology , COVID-19/immunology , Endothelial Cells/physiology , Nervous System/virology , SARS-CoV-2/physiology , Animals , Endothelial Cells/virology , Humans , Immunity, Innate , Interferons/metabolism , Signal Transduction
12.
J Int Med Res ; 48(7): 300060520939746, 2020 Jul.
Article in English | MEDLINE | ID: covidwho-690568

ABSTRACT

The novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 infection is a serious global concern. Increased morbidity and mortality is associated with older age, male gender, cardiovascular disease, diabetes, and smoking. As COVID-19 spreads from coastal borders, both state to state and country to country, our understanding of its pathophysiology has evolved. Age and type 2 diabetes mellitus (T2DM) play especially important roles in COVID-19 progression. T2DM is an age-related disease associated with metabolic syndrome, obesity, insulin resistance (hyperinsulinemia), hyperlipidemia, hypertension, hyperglycemia, and endothelial activation and dysfunction. This review evaluates the relationships and intersection between endothelial cell activation and dysfunction in T2DM and COVID-19. COVID-19 induces multiple injuries of the terminal bronchioles and alveolar blood-gas barrier and associated ultrastructural tissue remodeling. COVID-19 may unmask multiple vulnerabilities associated with T2DM including damage to the endothelial glycocalyx and multiple end-organ macro and microvascular diseases. Unmasking existing vulnerabilities in diabetic patients with COVID-19 is important. Globally, we must come together to better understand why T2DM is associated with increased COVID-19 morbidity and mortality.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Coronavirus Infections/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Endothelial Cells/physiology , Metabolic Syndrome/complications , Metabolic Syndrome/physiopathology , Pneumonia, Viral/complications , Pneumonia, Viral/physiopathology , Animals , Blood-Air Barrier/pathology , Blood-Air Barrier/physiopathology , Bronchioles/pathology , Bronchioles/physiopathology , COVID-19 , Comorbidity , Coronavirus Infections/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Drug Repositioning , Endothelial Cells/pathology , Humans , Metabolic Syndrome/epidemiology , Models, Biological , Pandemics , Pneumonia, Viral/epidemiology , Pulmonary Alveoli/physiology , Pulmonary Alveoli/physiopathology , Rats , SARS-CoV-2 , Wound Healing/physiology
13.
Crit Care ; 24(1): 365, 2020 06 19.
Article in English | MEDLINE | ID: covidwho-608395

ABSTRACT

In this viewpoint, we summarize the relevance of thromboinflammation in COVID-19 and discuss potential mechanisms of endothelial injury as a key point for the development of lung and distant organ dysfunction, with a focus on direct viral infection and cytokine-mediated injury. Entanglement between inflammation and coagulation and resistance to heparin provide a rationale to consider other therapeutic approaches in order to preserve endothelial function and limit microthrombosis, especially in severe forms. These strategies include nebulized heparin, N-acetylcysteine, plasma exchange and/or fresh frozen plasma, plasma derivatives to increase the level of endogenous anticoagulants (tissue factor pathway inhibitor, activated protein C, thrombomodulin, antithrombin), dipyridamole, complement blockers, different types of stem cells, and extracellular vesicles. An integrated therapy including these drugs has the potential to improve outcomes in COVID-19.


Subject(s)
Coronavirus Infections/therapy , Endothelial Cells/physiology , Inflammation/prevention & control , Pneumonia, Viral/therapy , Thrombosis/prevention & control , COVID-19 , Coronavirus Infections/physiopathology , Humans , Pandemics , Pneumonia, Viral/physiopathology
14.
Kidney Int ; 98(2): 314-322, 2020 08.
Article in English | MEDLINE | ID: covidwho-343018

ABSTRACT

The novel coronavirus disease COVID-19 originates in the lungs, but it may extend to other organs, causing, in severe cases, multiorgan damage, including cardiac injury and acute kidney injury. In severe cases, the presence of kidney injury is associated with increased risk of death, highlighting the relevance of this organ as a target of SARS-CoV-2 infection. COVID-19-associated tissue injury is not primarily mediated by viral infection, but rather is a result of the inflammatory host immune response, which drives hypercytokinemia and aggressive inflammation that affect lung parenchymal cells, diminishing oxygen uptake, but also endothelial cells, resulting in endotheliitis and thrombotic events and intravascular coagulation. The complement system represents the first response of the host immune system to SARS-CoV-2 infection, but there is growing evidence that unrestrained activation of complement induced by the virus in the lungs and other organs plays a major role in acute and chronic inflammation, endothelial cell dysfunction, thrombus formation, and intravascular coagulation, and ultimately contributes to multiple organ failure and death. In this review, we discuss the relative role of the different complement activation products in the pathogenesis of COVID-19-associated tissue inflammation and thrombosis and propose the hypothesis that blockade of the terminal complement pathway may represent a potential therapeutic option for the prevention and treatment of lung and multiorgan damage.


Subject(s)
Betacoronavirus , Complement Activation , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Animals , COVID-19 , Complement C5a/physiology , Coronavirus Infections/complications , Endothelial Cells/physiology , Humans , Inflammation/etiology , Mannose-Binding Lectin/physiology , Mice , Pandemics , Pneumonia, Viral/complications , SARS-CoV-2 , Thrombosis/etiology , Vascular Diseases/etiology
SELECTION OF CITATIONS
SEARCH DETAIL