Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Viruses ; 16(4)2024 04 22.
Article in English | MEDLINE | ID: mdl-38675987

ABSTRACT

Consistent with the biochemistry of coronaviruses as well established over decades, SARS-CoV-2 makes its initial attachment to host cells through the binding of its spike protein (SP) to sialylated glycans (containing the monosaccharide sialic acid) on the cell surface. The virus can then slide over and enter via ACE2. SARS-CoV-2 SP attaches particularly tightly to the trillions of red blood cells (RBCs), platelets and endothelial cells in the human body, each cell very densely coated with sialic acid surface molecules but having no ACE2 or minimal ACE2. These interlaced attachments trigger the blood cell aggregation, microvascular occlusion and vascular damage that underlie the hypoxia, blood clotting and related morbidities of severe COVID-19. Notably, the two human betacoronaviruses that express a sialic acid-cleaving enzyme are benign, while the other three-SARS, SARS-CoV-2 and MERS-are virulent. RBC aggregation experimentally induced in several animal species using an injected polysaccharide caused most of the same morbidities of severe COVID-19. This glycan biochemistry is key to disentangling controversies that have arisen over the efficacy of certain generic COVID-19 treatment agents and the safety of SP-based COVID-19 vaccines. More broadly, disregard for the active physiological role of RBCs yields unreliable or erroneous reporting of pharmacokinetic parameters as routinely obtained for most drugs and other bioactive agents using detection in plasma, with whole-blood levels being up to 30-fold higher. Appreciation of the active role of RBCs can elucidate the microvascular underpinnings of other health conditions, including cardiovascular disease, and therapeutic opportunities to address them.


Subject(s)
COVID-19 , Polysaccharides , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19/metabolism , COVID-19/virology , SARS-CoV-2/metabolism , Polysaccharides/metabolism , Animals , Spike Glycoprotein, Coronavirus/metabolism , Betacoronavirus/metabolism , Coronavirus Infections/metabolism , Erythrocytes/metabolism , Erythrocytes/virology , Pandemics , Microvessels/metabolism , Microvessels/virology , Virus Attachment , COVID-19 Drug Treatment , Endothelial Cells/metabolism , Endothelial Cells/virology , Angiotensin-Converting Enzyme 2/metabolism , Erythrocyte Aggregation
2.
Cell ; 185(3): 493-512.e25, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35032429

ABSTRACT

Severe COVID-19 is linked to both dysfunctional immune response and unrestrained immunopathology, and it remains unclear whether T cells contribute to disease pathology. Here, we combined single-cell transcriptomics and single-cell proteomics with mechanistic studies to assess pathogenic T cell functions and inducing signals. We identified highly activated CD16+ T cells with increased cytotoxic functions in severe COVID-19. CD16 expression enabled immune-complex-mediated, T cell receptor-independent degranulation and cytotoxicity not found in other diseases. CD16+ T cells from COVID-19 patients promoted microvascular endothelial cell injury and release of neutrophil and monocyte chemoattractants. CD16+ T cell clones persisted beyond acute disease maintaining their cytotoxic phenotype. Increased generation of C3a in severe COVID-19 induced activated CD16+ cytotoxic T cells. Proportions of activated CD16+ T cells and plasma levels of complement proteins upstream of C3a were associated with fatal outcome of COVID-19, supporting a pathological role of exacerbated cytotoxicity and complement activation in COVID-19.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Complement Activation , Proteome , SARS-CoV-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Transcriptome , Adult , Aged , Aged, 80 and over , COVID-19/virology , Chemotactic Factors/metabolism , Cytotoxicity, Immunologic , Endothelial Cells/virology , Female , Humans , Lymphocyte Activation , Male , Microvessels/virology , Middle Aged , Monocytes/metabolism , Neutrophils/metabolism , Receptors, IgG/metabolism , Single-Cell Analysis , Young Adult
3.
Cells ; 10(9)2021 08 31.
Article in English | MEDLINE | ID: mdl-34571912

ABSTRACT

COVID-19 presents with a wide range of clinical neurological manifestations. It has been recognized that SARS-CoV-2 infection affects both the central and peripheral nervous system, leading to smell and taste disturbances; acute ischemic and hemorrhagic cerebrovascular disease; encephalopathies and seizures; and causes most surviving patients to have long lasting neurological symptoms. Despite this, typical neuropathological features associated with the infection have still not been identified. Studies of post-mortem examinations of the cerebral cortex are obtained with difficulty due to laboratory safety concerns. In addition, they represent cases with different neurological symptoms, age or comorbidities, thus a larger number of brain autoptic data from multiple institutions would be crucial. Histopathological findings described here are aimed to increase the current knowledge on neuropathology of COVID-19 patients. We report post-mortem neuropathological findings of ten COVID-19 patients. A wide range of neuropathological lesions were seen. The cerebral cortex of all patients showed vascular changes, hyperemia of the meninges and perivascular inflammation in the cerebral parenchyma with hypoxic neuronal injury. Perivascular lymphocytic inflammation of predominantly CD8-positive T cells mixed with CD68-positive macrophages, targeting the disrupted vascular wall in the cerebral cortex, cerebellum and pons were seen. Our findings support recent reports highlighting a role of microvascular injury in COVID-19 neurological manifestations.


Subject(s)
COVID-19/pathology , Cerebral Cortex/pathology , Aged , Aged, 80 and over , Autopsy , Brain/pathology , Brain/virology , Brain Diseases/pathology , Brain Diseases/virology , CD8-Positive T-Lymphocytes/pathology , Cerebral Cortex/virology , Female , Humans , Inflammation , Macrophages/pathology , Male , Microvessels/pathology , Microvessels/virology , Middle Aged , Nervous System Diseases/pathology , Nervous System Diseases/virology , SARS-CoV-2/pathogenicity
4.
Physiol Rep ; 9(3): e14726, 2021 02.
Article in English | MEDLINE | ID: mdl-33523608

ABSTRACT

Corona virus disease 2019 (COVID-19) causes symptoms from multiple organs after infection by severe acute respiratory syndrome corona virus 2 (SARS CoV-2). They range from early, low blood oxygen levels (hypoxemia) without breathlessness ("silent hypoxia"), delirium, rashes, and loss of smell (anosmia), to persisting chest pain, muscle weakness and -pain, fatigue, confusion, memory problems and difficulty to concentrate ("brain fog"), mood changes, and unexpected onset of hypertension or diabetes. SARS CoV-2 affects the microcirculation, causing endothelial cell swelling and damage (endotheliitis), microscopic blood clots (microthrombosis), capillary congestion, and damage to pericytes that are integral to capillary integrity and barrier function, tissue repair (angiogenesis), and scar formation. Similar to other instances of critical illness, COVID-19 is also associated with elevated cytokine levels in the systemic circulation. This review examines how capillary damage and inflammation may contribute to these acute and persisting COVID-19 symptoms by interfering with blood and tissue oxygenation and with brain function. Undetectable by current diagnostic methods, capillary flow disturbances limit oxygen diffusion exchange in lungs and tissue and may therefore cause hypoxemia and tissue hypoxia. The review analyzes the combined effects of COVID-19-related capillary damage, pre-existing microvascular changes, and upstream vascular tone on tissue oxygenation in key organs. It identifies a vicious cycle, as infection- and hypoxia-related inflammation cause capillary function to deteriorate, which in turn accelerates hypoxia-related inflammation and tissue damage. Finally, the review addresses the effects of low oxygen and high cytokine levels in brain tissue on neurotransmitter synthesis and mood. Methods to assess capillary functions in human organs and therapeutic means to protect capillary functions and stimulate capillary bed repair may prove important for the individualized management of COVID-19 patients and targeted rehabilitation strategies.


Subject(s)
COVID-19/complications , Microvessels/pathology , Oxygen Consumption , Oxygen/metabolism , Animals , COVID-19/metabolism , COVID-19/pathology , Humans , Inflammation , Microvessels/metabolism , Microvessels/virology , Oxygen/blood , SARS-CoV-2/pathogenicity , Post-Acute COVID-19 Syndrome
5.
Cells ; 10(2)2021 02 10.
Article in English | MEDLINE | ID: mdl-33578631

ABSTRACT

Clinical manifestations of coronavirus disease 2019 (COVID-19) in pregnant women are diverse, and little is known of the impact of the disease on placental physiology. Severe acute respiratory syndrome coronavirus (SARS-CoV-2) has been detected in the human placenta, and its binding receptor ACE2 is present in a variety of placental cells, including endothelium. Here, we analyze the impact of COVID-19 in placental endothelium, studying by immunofluorescence the expression of von Willebrand factor (vWf), claudin-5, and vascular endothelial (VE) cadherin in the decidua and chorionic villi of placentas from women with mild and severe COVID-19 in comparison to healthy controls. Our results indicate that: (1) vWf expression increases in the endothelium of decidua and chorionic villi of placentas derived from women with COVID-19, being higher in severe cases; (2) Claudin-5 and VE-cadherin expression decrease in the decidua and chorionic villus of placentas from women with severe COVID-19 but not in those with mild disease. Placental histological analysis reveals thrombosis, infarcts, and vascular wall remodeling, confirming the deleterious effect of COVID-19 on placental vessels. Together, these results suggest that placentas from women with COVID-19 have a condition of leaky endothelium and thrombosis, which is sensitive to disease severity.


Subject(s)
COVID-19/complications , Placenta/blood supply , Placenta/pathology , Pregnancy Complications, Cardiovascular/etiology , Pregnancy Complications, Infectious/etiology , Thrombosis/etiology , Adult , Antigens, CD/analysis , COVID-19/pathology , COVID-19/virology , Cadherins/analysis , Claudin-5/analysis , Endothelium/blood supply , Endothelium/pathology , Endothelium/virology , Female , Humans , Infant, Newborn , Microvessels/pathology , Microvessels/virology , Pregnancy , Pregnancy Complications, Cardiovascular/pathology , Pregnancy Complications, Cardiovascular/virology , Pregnancy Complications, Infectious/pathology , Pregnancy Complications, Infectious/virology , SARS-CoV-2/isolation & purification , Thrombosis/pathology , Thrombosis/virology , Young Adult , von Willebrand Factor/analysis
7.
Ann Diagn Pathol ; 51: 151682, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33360731

ABSTRACT

Neurologic complications of symptomatic COVID-19 are common. Brain tissues from 13 autopsies of people who died of COVID-19 were examined. Cultured endothelial and neuronal cells were incubated with and wild type mice were injected IV with different spike subunits. In situ analyses were used to detect SARS-CoV-2 proteins and the host response. In 13/13 brains from fatal COVID-19, pseudovirions (spike, envelope, and membrane proteins without viral RNA) were present in the endothelia of microvessels ranging from 0 to 14 positive cells/200× field (mean 4.3). The pseudovirions strongly co-localized with caspase-3, ACE2, IL6, TNFα, and C5b-9. The surrounding neurons demonstrated increased NMDAR2 and neuronal NOS plus decreased MFSD2a and SHIP1 proteins. Tail vein injection of the full length S1 spike subunit in mice led to neurologic signs (increased thirst, stressed behavior) not evident in those injected with the S2 subunit. The S1 subunit localized to the endothelia of microvessels in the mice brain and showed co-localization with caspase-3, ACE2, IL6, TNFα, and C5b-9. The surrounding neurons showed increased neuronal NOS and decreased MFSD2a. It is concluded that ACE2+ endothelial damage is a central part of SARS-CoV2 pathology and may be induced by the spike protein alone. Thus, the diagnostic pathologist can use either hematoxylin and eosin stain or immunohistochemistry for caspase 3 and ACE2 to document the endothelial cell damage of COVID-19.


Subject(s)
COVID-19/virology , Endothelial Cells/virology , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Adult , Aged , Aged, 80 and over , Animals , Autopsy/methods , Disease Models, Animal , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Microvessels/metabolism , Microvessels/virology , Middle Aged , Protein Subunits/metabolism , RNA, Viral/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics
8.
Ann Diagn Pathol ; 50: 151645, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33248385

ABSTRACT

The objective of this study was to elucidate the pathophysiology that underlies severe COVID-19 by assessing the histopathology and the in situ detection of infectious SARS-CoV-2 and viral capsid proteins along with the cellular target(s) and host response from twelve autopsies. There were three key findings: 1) high copy infectious virus was limited mostly to the alveolar macrophages and endothelial cells of the septal capillaries; 2) viral spike protein without viral RNA localized to ACE2+ endothelial cells in microvessels that were most abundant in the subcutaneous fat and brain; 3) although both infectious virus and docked viral spike protein was associated with complement activation, only the endocytosed pseudovirions induced a marked up-regulation of the key COVID-19 associated proteins IL6, TNF alpha, IL1 beta, p38, IL8, and caspase 3. Importantly, this microvasculitis was associated with characteristic findings on hematoxylin and eosin examination that included endothelial degeneration and resultant basement membrane zone disruption and reduplication. It is concluded that serious COVID-19 infection has two distinct mechanisms: 1) a microangiopathy of pulmonary capillaries associated with a high infectious viral load where endothelial cell death releases pseudovirions into the circulation, and 2) the pseudovirions dock on ACE2+ endothelial cells most prevalent in the skin/subcutaneous fat and brain that activates the complement pathway/coagulation cascade resulting in a systemic procoagulant state as well as the expression of cytokines that produce the cytokine storm. The data predicts a favorable response to therapies based on either removal of circulating viral proteins and/or blunting of the endothelial-induced response.


Subject(s)
COVID-19/physiopathology , Capsid Proteins/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Thrombotic Microangiopathies/physiopathology , Vascular Diseases/physiopathology , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/metabolism , Autopsy , COVID-19/virology , Capsid Proteins/genetics , Endothelial Cells/enzymology , Endothelial Cells/virology , Female , Humans , Lung/physiopathology , Lung/virology , Male , Microvessels/physiopathology , Microvessels/virology , Middle Aged , RNA, Viral/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Thrombotic Microangiopathies/virology , Vascular Diseases/virology , Virion
9.
Front Endocrinol (Lausanne) ; 11: 596898, 2020.
Article in English | MEDLINE | ID: mdl-33281748

ABSTRACT

Increasing evidence demonstrated that the expression of Angiotensin I-Converting Enzyme type 2 (ACE2) is a necessary step for SARS-CoV-2 infection permissiveness. In light of the recent data highlighting an association between COVID-19 and diabetes, a detailed analysis aimed at evaluating ACE2 expression pattern distribution in human pancreas is still lacking. Here, we took advantage of INNODIA network EUnPOD biobank collection to thoroughly analyze ACE2, both at mRNA and protein level, in multiple human pancreatic tissues and using several methodologies. Using multiple reagents and antibodies, we showed that ACE2 is expressed in human pancreatic islets, where it is preferentially expressed in subsets of insulin producing ß-cells. ACE2 is also highly expressed in pancreas microvasculature pericytes and moderately expressed in rare scattered ductal cells. By using different ACE2 antibodies we showed that a recently described short-ACE2 isoform is also prevalently expressed in human ß-cells. Finally, using RT-qPCR, RNA-seq and High-Content imaging screening analysis, we demonstrated that pro-inflammatory cytokines, but not palmitate, increase ACE2 expression in the ß-cell line EndoC-ßH1 and in primary human pancreatic islets. Taken together, our data indicate a potential link between SARS-CoV-2 and diabetes through putative infection of pancreatic microvasculature and/or ductal cells and/or through direct ß-cell virus tropism.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Insulin-Secreting Cells/metabolism , Microvessels/metabolism , Pancreas/metabolism , SARS-CoV-2/isolation & purification , COVID-19/metabolism , COVID-19/pathology , Cells, Cultured , Cytokines/metabolism , Humans , Insulin-Secreting Cells/virology , Microvessels/virology , Pancreas/virology
10.
Hum Pathol ; 106: 106-116, 2020 12.
Article in English | MEDLINE | ID: mdl-33058948

ABSTRACT

The purpose of this study was to examine the deltoid skin biopsy in twenty-three patients with coronavirus disease 2019 (COVID-19), most severely ill, for vascular complement deposition and correlate this with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral RNA and protein localization and ACE2 expression. Deltoid skin microvascular complement screening has been applied to patients with various systemic complement-mediated microvascular syndromes, best exemplified by atypical hemolytic uremic syndrome. In 21 of 23 cases, substantial microvascular deposition of complement components was identified. The two patients without significant complement deposition included one patient with moderate disease and a severely ill patient who although on a ventilator for a day was discharged after 3 days. The dominant microvascular complement immunoreactant identified was the terminal membranolytic attack complex C5b-9. Microvascular complement deposition strongly colocalized in situ with the SARS-CoV-2 viral proteins including spike glycoproteins in the endothelial cells as well as the viral receptor ACE2 in lesional and nonlesional skin; viral RNA was not evident. Microvascular SARS-CoV-2 viral protein, complement, and ACE2 expression was most conspicuous in the subcutaneous fat. Although the samples from severely ill patients with COVID-19 were from grossly normal skin, light microscopically focal microvascular abnormalities were evident that included endothelial cell denudement, basement membrane zone reduplication, and small thrombi. It is concluded that complement activation is common in grossly normal skin, especially in the subcutaneous fat which may provide a link between severe disease and obesity, in people with severe COVID-19, and the strong colocalization with the ACE2 receptor and viral capsid proteins without viral RNA suggests that circulating viral proteins (ie, pseudovirions) may dock onto the endothelial of these microvessels and induce complement activation.


Subject(s)
COVID-19/virology , Endothelial Cells/virology , Microvessels/virology , SARS-CoV-2/pathogenicity , Adult , Aged , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , Complement Activation/immunology , Endothelial Cells/metabolism , Female , Humans , Male , Microvessels/metabolism , Middle Aged , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , RNA, Viral/genetics
11.
Heart Lung Circ ; 29(11): 1596-1602, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32972810

ABSTRACT

The recently described severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected millions of people, with thousands of fatalities. It has prompted global efforts in research, with focus on the pathophysiology of coronavirus disease-19 (COVID-19), and a rapid surge of publications. COVID-19 has been associated with a myriad of clinical manifestations, including the lungs, heart, kidneys, central nervous system, gastrointestinal system, skin, and blood coagulation abnormalities. The endothelium plays a key role in organ dysfunction associated with severe infection, and current data suggest that it is also involved in SARS-CoV-2-induced sepsis. This critical review aimed to address a possible unifying mechanism underlying the diverse complications of COVID-19: microvascular dysfunction, with emphasis on the renin-angiotensin system. In addition, research perspectives are suggested in order to expand understanding of the pathophysiology of the infection.


Subject(s)
Coronavirus Infections , Microvessels , Pandemics , Pneumonia, Viral , Renin-Angiotensin System/physiology , Betacoronavirus/physiology , COVID-19 , Coronavirus Infections/metabolism , Coronavirus Infections/physiopathology , Humans , Microvessels/metabolism , Microvessels/physiopathology , Microvessels/virology , Pneumonia, Viral/metabolism , Pneumonia, Viral/physiopathology , SARS-CoV-2
12.
Int J Mol Sci ; 21(15)2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32722052

ABSTRACT

Microvascular endothelial cells constitute potential targets for exogenous microorganisms, in particular for vector-borne pathogens. Their phenotypic and functional variations according to the organs they are coming from provide an explanation of the organ selectivity expressed in vivo by pathogens. In order to make available relevant tools for in vitro studies of infection mechanisms, our aim was to immortalize bovine organospecific endothelial cells but also to assess their permissivity to viral infection. Using transfection with SV40 large T antigen, six bovine microvascular endothelial cell lines from various organs and one macrovascular cell line from an umbilical cord were established. They display their own panel of endothelial progenitor/mature markers, as assessed by flow cytometry and RT-qPCR, as well as the typical angiogenesis capacity. Using both Bluetongue and foot-and-mouth disease viruses, we demonstrate that some cell lines are preferentially infected. In addition, they can be transfected and are able to express viral proteins such as BTV8-NS3. Such microvascular endothelial cell lines bring innovative tools for in vitro studies of infection by viruses or bacteria, allowing for the study of host-pathogen interaction mechanisms with the actual in vivo target cells. They are also suitable for applications linked to microvascularization, such as anti-angiogenic and anti-tumor research, growing fields in veterinary medicine.


Subject(s)
Endothelial Cells/metabolism , Microvessels/metabolism , Models, Biological , Virus Diseases , Animals , Cattle , Cell Line , Endothelial Cells/pathology , Endothelial Cells/virology , Microvessels/pathology , Microvessels/virology
14.
Clin Chim Acta ; 507: 167-173, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32348783

ABSTRACT

Early clinical evidence suggests that severe cases of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), are frequently characterized by hyperinflammation, imbalance of renin-angiotensin-aldosterone system, and a particular form of vasculopathy, thrombotic microangiopathy, and intravascular coagulopathy. In this paper, we present an immunothrombosis model of COVID-19. We discuss the underlying pathogenesis and the interaction between multiple systems, resulting in propagation of immunothrombosis, which through investigation in the coming weeks, may lead to both an improved understanding of COVID-19 pathophysiology and identification of innovative and efficient therapeutic targets to reverse the otherwise unfavorable clinical outcome of many of these patients.


Subject(s)
Aldosterone/immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Microvessels/immunology , Pneumonia, Viral/immunology , Renin-Angiotensin System/immunology , Thrombophilia/immunology , Thrombosis/immunology , COVID-19 , Coronavirus Infections/physiopathology , Humans , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/physiopathology , Inflammation/virology , Microvessels/physiopathology , Microvessels/virology , Pandemics , Pneumonia, Viral/physiopathology , SARS-CoV-2 , Thrombophilia/physiopathology , Thrombophilia/virology , Thrombosis/physiopathology , Thrombosis/virology
15.
Transl Res ; 220: 1-13, 2020 06.
Article in English | MEDLINE | ID: mdl-32299776

ABSTRACT

Acute respiratory failure and a systemic coagulopathy are critical aspects of the morbidity and mortality characterizing infection with severe acute respiratory distress syndrome-associated coronavirus-2, the etiologic agent of Coronavirus disease 2019 (COVID-19). We examined skin and lung tissues from 5 patients with severe COVID-19 characterized by respiratory failure (n= 5) and purpuric skin rash (n = 3). COVID-19 pneumonitis was predominantly a pauci-inflammatory septal capillary injury with significant septal capillary mural and luminal fibrin deposition and permeation of the interalveolar septa by neutrophils. No viral cytopathic changes were observed and the diffuse alveolar damage (DAD) with hyaline membranes, inflammation, and type II pneumocyte hyperplasia, hallmarks of classic acute respiratory distress syndrome, were not prominent. These pulmonary findings were accompanied by significant deposits of terminal complement components C5b-9 (membrane attack complex), C4d, and mannose binding lectin (MBL)-associated serine protease (MASP)2, in the microvasculature, consistent with sustained, systemic activation of the complement pathways. The purpuric skin lesions similarly showed a pauci-inflammatory thrombogenic vasculopathy, with deposition of C5b-9 and C4d in both grossly involved and normally-appearing skin. In addition, there was co-localization of COVID-19 spike glycoproteins with C4d and C5b-9 in the interalveolar septa and the cutaneous microvasculature of 2 cases examined. In conclusion, at least a subset of sustained, severe COVID-19 may define a type of catastrophic microvascular injury syndrome mediated by activation of complement pathways and an associated procoagulant state. It provides a foundation for further exploration of the pathophysiologic importance of complement in COVID-19, and could suggest targets for specific intervention.


Subject(s)
Betacoronavirus , Complement System Proteins/metabolism , Coronavirus Infections/complications , Microvessels/pathology , Pneumonia, Viral/complications , Respiratory Insufficiency/etiology , Thrombosis/etiology , Adult , Aged , COVID-19 , Complement Activation/physiology , Coronavirus Infections/pathology , Female , Humans , Male , Microvessels/virology , Middle Aged , Pandemics , Pneumonia, Viral/pathology , Purpura/etiology , Purpura/pathology , Purpura/virology , Respiratory Insufficiency/pathology , SARS-CoV-2 , Thrombosis/pathology
16.
Anat Rec (Hoboken) ; 302(5): 818-824, 2019 05.
Article in English | MEDLINE | ID: mdl-30312024

ABSTRACT

Vascular damage has been reported to contribute to atresia formation in several diseases including biliary atresia. This study focused on the extrahepatic biliary plexus in experimental biliary atresia. Newborn BALB/cAnNCrl-pups were infected with rhesus rotavirus within 24 hr after birth to induce experimental biliary atresia. The extrahepatic biliary plexus was examined by confocal microscopy on whole-mount preparations, scored by three independent researchers, and further evaluated at the subcellular level with transmission electron microscopy. Imaging results revealed a progressive destruction of the extrahepatic biliary vascular plexus in the course of experimental biliary atresia induced by rotavirus infection. Endothelial cell damage was already visible as cell swelling and necrosis in the first days after infection and a damaged microcirculation that rapidly deteriorated with progression of obliterative cholangiopathy, was observed in the infected mice as early as 72 hr after birth. In experimental biliary atresia, the destruction of the extrahepatic biliary vascular plexus starts already in the first days postinfection and clearly precedes the morphological symptoms of atresia. The deterioration of the vascular bed architecture continues with disease progression. Therefore, we conclude that the (ultra)structural changes in the extrahepatic biliary microvasculature occurring before the visible onset of atresia has a predictive diagnostic value and this impairment in blood supply to the extrahepatic bile duct may be an important contributing factor to the pathogenesis of acquired biliary atresia. Anat Rec, 2018. © 2018 Wiley Periodicals, Inc. Anat Rec, 302:818-824, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
Bile Ducts, Extrahepatic/blood supply , Biliary Atresia/pathology , Microvessels/pathology , Rotavirus Infections/pathology , Rotavirus/pathogenicity , Animals , Animals, Newborn , Bile Ducts, Extrahepatic/pathology , Bile Ducts, Extrahepatic/virology , Biliary Atresia/virology , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Microscopy, Electron, Transmission , Microvessels/ultrastructure , Microvessels/virology , Rotavirus Infections/virology
17.
Arch Immunol Ther Exp (Warsz) ; 67(1): 27-40, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30238127

ABSTRACT

Diseases caused by dengue virus (DENV) are a major public health problem worldwide, considered one of the infections with more prevalence in tropical and subtropical zones of the world. Despite the intense research in the pathogenesis of DENV, this feature is not well understood. One of the main target cells for DENV infection is monocytes; these phagocytes can play a dual role, since they are essential to control viremia, but they also participate in the induction of tissue damage during DENV infection. Monocytes produce different pro-inflammatory cytokines and chemokines in response to infection, and also mediate endothelial damage. In peripheral blood, monocytes can be divided into three different subpopulations, namely classical, intermediate and non-classical, which differ in frequency, cytokine production, among others. Studies in the last years suggest that non-classical monocytes have higher affinity for microvasculature endothelium compared to other type of monocytes, which implies that they could be more involved in the increase of endothelial permeability observed during DENV infection. This review provides a general view of the role of monocytes and their subpopulations in DENV pathogenesis and its effect in viral replication. Finally, the potential contribution of these phagocytes in the alterations of endothelial permeability is discussed.


Subject(s)
Dengue Virus/pathogenicity , Dengue/virology , Monocytes/virology , Animals , Capillary Permeability , Cytokines/immunology , Cytokines/metabolism , Dengue/immunology , Dengue/metabolism , Dengue Virus/growth & development , Dengue Virus/immunology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/virology , Host-Pathogen Interactions , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Microvessels/immunology , Microvessels/metabolism , Microvessels/virology , Monocytes/immunology , Monocytes/metabolism , Phagocytosis , Signal Transduction , Virus Replication
18.
Fluids Barriers CNS ; 15(1): 15, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29759080

ABSTRACT

Zika virus (ZIKV) is a flavivirus that is highly neurotropic causing congenital abnormalities and neurological damage to the central nervous systems (CNS). In this study, we used a human induced pluripotent stem cell (iPSC)-derived blood brain barrier (BBB) model to demonstrate that ZIKV can infect brain endothelial cells (i-BECs) without compromising the BBB barrier integrity or permeability. Although no disruption to the BBB was observed post-infection, ZIKV particles were released on the abluminal side of the BBB model and infected underlying iPSC-derived neural progenitor cells (i-NPs). AXL, a putative ZIKV cellular entry receptor, was also highly expressed in ZIKV-susceptible i-BEC and i-NPs. This iPSC-derived BBB model can help elucidate the mechanism by which ZIKV can infect BECs, cross the BBB and gain access to the CNS.


Subject(s)
Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Zika Virus/metabolism , Capillary Permeability/physiology , Cell Culture Techniques , Endothelial Cells/metabolism , Endothelial Cells/virology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/virology , Microvessels/metabolism , Microvessels/virology , Zika Virus Infection/metabolism , Zika Virus Infection/virology
19.
J Alzheimers Dis ; 60(2): 359-369, 2017.
Article in English | MEDLINE | ID: mdl-28800332

ABSTRACT

The polymicrobial dysbiotic subgingival biofilm microbes associated with periodontal disease appear to contribute to developing pathologies in distal body sites, including the brain. This study examined oxidative stress, in the form of increased protein carbonylation and oxidative protein damage, in the tumor necrosis factor-α (TNF-α) transgenic mouse that models inflammatory TNF-α excess during bacterial infection; and in the apolipoprotein knockout (ApoE-/-) mouse brains, following Porphyromonas gingivalis gingival monoinfection. Following 2,4-dinitrophenylhydrazine derivatization, carbonyl groups were detected in frontal lobe brain tissue lysates by immunoblotting and immunohistochemical analysis of fixed tissue sections from the frontotemporal lobe and the hippocampus. Immunoblot analysis confirmed the presence of variable carbonyl content and oxidative protein damage in all lysates, with TNF-α transgenic blots exhibiting increased protein carbonyl content, with consistently prominent bands at 25 kDa (p = 0.0001), 43 kDa, and 68 kDa, over wild-type mice. Compared to sham-infected ApoE-/- mouse blots, P. gingivalis-infected brain tissue blots demonstrated the greatest detectable protein carbonyl content overall, with numerous prominent bands at 25 kDa (p = 0.001) and 43 kDa (p = 0.0001) and an exclusive band to this group between 30-43 kDa* (p = 0.0001). In addition, marked immunostaining was detected exclusively in the microvasculature in P. gingivalis-infected hippocampal tissue sections, compared to sham-infected, wild-type, and TNF-α transgenic mice. This study revealed that the hippocampal microvascular structure of P. gingivalis-infected ApoE-/- mice possesses elevated oxidative stress levels, resulting in the associated tight junction proteins being susceptible to increased oxidative/proteolytic degradation, leading to a loss of functional integrity.


Subject(s)
Apolipoproteins E/deficiency , Bacteroidaceae Infections/physiopathology , Microvessels/pathology , Oxidative Stress/genetics , Porphyromonas gingivalis/pathogenicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Apolipoproteins E/genetics , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/virology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/virology , Phenylhydrazines/metabolism , Protein Carbonylation/genetics , Protein Carbonylation/physiology , Tumor Necrosis Factor-alpha/genetics
20.
mBio ; 8(4)2017 07 11.
Article in English | MEDLINE | ID: mdl-28698279

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne Flavivirus that has emerged as the cause of encephalitis and fetal microencephaly in the Americas. ZIKV uniquely persists in human bodily fluids for up to 6 months, is sexually transmitted, and traverses the placenta and the blood-brain barrier (BBB) to damage neurons. Cells that support persistent ZIKV replication and mechanisms by which ZIKV establishes persistence remain enigmatic but central to ZIKV entry into protected neuronal compartments. The endothelial cell (EC) lining of capillaries normally constrains transplacental transmission and forms the BBB, which selectively restricts access of blood constituents to neurons. We found that ZIKV (strain PRVABC59) persistently infects and continuously replicates in primary human brain microvascular ECs (hBMECs), without cytopathology, for >9 days and following hBMEC passage. ZIKV did not permeabilize hBMECs but was released basolaterally from polarized hBMECs, suggesting a direct mechanism for ZIKV to cross the BBB. ZIKV-infected hBMECs were rapidly resistant to alpha interferon (IFN-α) and transiently induced, but failed to secrete, IFN-ß and IFN-λ. Global transcriptome analysis determined that ZIKV constitutively induced IFN regulatory factor 7 (IRF7), IRF9, and IFN-stimulated genes (ISGs) 1 to 9 days postinfection, despite persistently replicating in hBMECs. ZIKV constitutively induced ISG15, HERC5, and USP18, which are linked to hepatitis C virus (HCV) persistence and IFN regulation, chemokine CCL5, which is associated with immunopathogenesis, as well as cell survival factors. Our results reveal that hBMECs act as a reservoir of persistent ZIKV replication, suggest routes for ZIKV to cross hBMECs into neuronal compartments, and define novel mechanisms of ZIKV persistence that can be targeted to restrict ZIKV spread.IMPORTANCE ZIKV persists in patients, crossing placental and neuronal barriers, damaging neurons, and causing fetal microencephaly. We found that ZIKV persistently infects brain endothelial cells that normally protect neurons from viral exposure. hBMECs are not damaged by ZIKV infection and, analogous to persistent HCV infection, ZIKV constitutively induces and evades antiviral ISG and IFN responses to continuously replicate in hBMECs. As a result, hBMECs provide a protective niche for systemic ZIKV spread and a viral reservoir localized in the normally protective blood-brain barrier. Consistent with the spread of ZIKV into neuronal compartments, ZIKV was released basolaterally from hBMECs. Our findings define hBMEC responses that contribute to persistent ZIKV infection and potential targets for clearing ZIKV infections from hBMECs. These results further suggest roles for additional ZIKV-infected ECs to facilitate viral spread and persistence in the protected placental, retinal, and testicular compartments.


Subject(s)
Brain/blood supply , Endothelial Cells/virology , Microvessels/virology , Virus Release , Virus Replication , Zika Virus/physiology , Antiviral Agents/pharmacology , Brain/virology , Cells, Cultured , Chemokine CCL5/genetics , Endopeptidases/genetics , Endothelial Cells/drug effects , Endothelial Cells/pathology , Gene Expression Profiling , Humans , Immunity, Innate , Interferon Regulatory Factor-7/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-alpha/pharmacology , Interferon-beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Microvessels/cytology , Ubiquitin Thiolesterase , Virus Internalization , Zika Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...