Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Clin Invest ; 131(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34060490

ABSTRACT

Worse outcomes occur in aged compared with young populations after infections with respiratory viruses, including pathogenic coronaviruses (SARS-CoV, MERS-CoV, and SARS-CoV-2), and are associated with a suboptimal lung milieu ("inflammaging"). We previously showed that a single inducible phospholipase, PLA2G2D, is associated with a proresolving/antiinflammatory response in the lungs, and increases with age. Survival was increased in naive Pla2g2d-/- mice infected with SARS-CoV resulting from augmented respiratory dendritic cell (rDC) activation and enhanced priming of virus-specific T cells. Here, in contrast, we show that intranasal immunization provided no additional protection in middle-aged Pla2g2d-/- mice infected with any of the 3 pathogenic human coronaviruses because virtually no virus-specific antibodies or follicular helper CD4+ T (Tfh) cells were produced. Using MERS-CoV-infected mice, we found that these effects did not result from T or B cell intrinsic factors. Rather, they resulted from enhanced, and ultimately, pathogenic rDC activation, as manifested most prominently by enhanced IL-1ß expression. Wild-type rDC transfer to Pla2g2d-/- mice in conjunction with partial IL-1ß blockade reversed this defect and resulted in increased virus-specific antibody and Tfh responses. Together, these results indicate that PLA2G2D has an unexpected role in the lungs, serving as an important modulator of rDC activation, with protective and pathogenic effects in respiratory coronavirus infections and immunization, respectively.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation , COVID-19/immunology , Group II Phospholipases A2/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , SARS-CoV-2/immunology , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Animals , COVID-19/enzymology , COVID-19/genetics , Chlorocebus aethiops , Group II Phospholipases A2/deficiency , Mice , Mice, Knockout , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/genetics , Vero Cells
2.
Infection ; 48(5): 665-669, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32737833

ABSTRACT

Novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) became pandemic by the end of March 2020. In contrast to the 2002-2003 SARS-CoV outbreak, which had a higher pathogenicity and lead to higher mortality rates, SARSCoV-2 infection appears to be much more contagious. Moreover, many SARS-CoV-2 infected patients are reported to develop low-titer neutralizing antibody and usually suffer prolonged illness, suggesting a more effective SARS-CoV-2 immune surveillance evasion than SARS-CoV. This paper summarizes the current state of art about the differences and similarities between the pathogenesis of the two coronaviruses, focusing on receptor binding domain, host cell entry and protease activation. Such differences may provide insight into possible intervention strategies to fight the pandemic.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/epidemiology , Pandemics , Pneumonia, Viral/epidemiology , Severe Acute Respiratory Syndrome/epidemiology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Spike Glycoprotein, Coronavirus/immunology , Angiotensin-Converting Enzyme 2 , Antibodies, Viral/biosynthesis , Betacoronavirus/immunology , COVID-19 , Cathepsins/genetics , Cathepsins/immunology , Coronavirus Infections/enzymology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Enzyme Activation/immunology , Humans , Immune Evasion , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/immunology , Pneumonia, Viral/enzymology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Protein Binding , Protein Domains , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2 , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/pathology , Severity of Illness Index , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Virus Internalization , Virus Replication
3.
SEMERGEN, Soc. Esp. Med. Rural Gen. (Ed. Impr.) ; 46(supl.1): 55-61, ago. 2020. tab, graf
Article in Spanish | IBECS | ID: ibc-192615

ABSTRACT

La pandemia por COVID-19 ha provocado un desajuste en todos los sistemas de salud. La mayoría de los países habían olvidado cómo comportarse ante una epidemia de estas características sin disponer de los recursos adecuados. Es preciso realizar un balance de todo lo sucedido, instruir a la población y generar un nuevo conocimiento que nos permita afrontar nuevas epidemias


The COVID-19 pandemic has caused a mismatch in all health systems. Most countries had forgotten how to behave in the face of such an epidemic without adequate resources. We need to take stock of everything that has happened, instruct the population and generate a new knowledge that allows us to face new epidemics


Subject(s)
Humans , Coronavirus Infections/epidemiology , Severe Acute Respiratory Syndrome/enzymology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Pandemics/prevention & control , Pandemics/statistics & numerical data , Communicable Disease Control/trends , 34661/methods , Surge Capacity/organization & administration
4.
Mol Syst Biol ; 16(7): e9841, 2020 07.
Article in English | MEDLINE | ID: mdl-32715628

ABSTRACT

Infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) leads to coronavirus disease 2019 (COVID-19), which poses an unprecedented worldwide health crisis, and has been declared a pandemic by the World Health Organization (WHO) on March 11, 2020. The angiotensin converting enzyme 2 (ACE2) has been suggested to be the key protein used by SARS-CoV-2 for host cell entry. In their recent work, Lindskog and colleagues (Hikmet et al, 2020) report that ACE2 is expressed at very low protein levels-if at all-in respiratory epithelial cells. Severe COVID-19, however, is characterized by acute respiratory distress syndrome and extensive damage to the alveoli in the lung parenchyma. Then, what is the role of the airway epithelium in the early stages of COVID-19, and which cells need to be studied to characterize the biological mechanisms responsible for the progression to severe disease after initial infection by the novel coronavirus?


Subject(s)
Coronavirus Infections/metabolism , Coronavirus Infections/virology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Severe Acute Respiratory Syndrome/metabolism , Severe Acute Respiratory Syndrome/virology , Angiotensin-Converting Enzyme 2 , Betacoronavirus , COVID-19 , Conjunctiva/metabolism , Coronavirus Infections/enzymology , Host Microbial Interactions/genetics , Humans , Organ Specificity , Pandemics , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/enzymology , SARS-CoV-2 , Severe Acute Respiratory Syndrome/enzymology , Spike Glycoprotein, Coronavirus/metabolism
5.
Leukemia ; 34(7): 1805-1815, 2020 07.
Article in English | MEDLINE | ID: mdl-32518419

ABSTRACT

A subgroup of patients with severe COVID-19 suffers from progression to acute respiratory distress syndrome and multiorgan failure. These patients present with progressive hyperinflammation governed by proinflammatory cytokines. An interdisciplinary COVID-19 work flow was established to detect patients with imminent or full blown hyperinflammation. Using a newly developed COVID-19 Inflammation Score (CIS), patients were prospectively stratified for targeted inhibition of cytokine signalling by the Janus Kinase 1/2 inhibitor ruxolitinib (Rux). Patients were treated with efficacy/toxicity guided step up dosing up to 14 days. Retrospective analysis of CIS reduction and clinical outcome was performed. Out of 105 patients treated between March 30th and April 15th, 2020, 14 patients with a CIS ≥ 10 out of 16 points received Rux over a median of 9 days with a median cumulative dose of 135 mg. A total of 12/14 patients achieved significant reduction of CIS by ≥25% on day 7 with sustained clinical improvement in 11/14 patients without short term red flag warnings of Rux-induced toxicity. Rux treatment for COVID-19 in patients with hyperinflammation is shown to be safe with signals of efficacy in this pilot case series for CRS-intervention to prevent or overcome multiorgan failure. A multicenter phase-II clinical trial has been initiated (NCT04338958).


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/drug therapy , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Pneumonia, Viral/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Severe Acute Respiratory Syndrome/drug therapy , Adult , Aged , Aged, 80 and over , Betacoronavirus/drug effects , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Clinical Trials as Topic , Coronavirus Infections/enzymology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cytokine Release Syndrome/enzymology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/immunology , Drug Administration Schedule , Female , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Inflammation , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Male , Middle Aged , Nitriles , Pandemics , Patient Safety , Pneumonia, Viral/enzymology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Pyrimidines , Retrospective Studies , SARS-CoV-2 , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/virology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Treatment Outcome
6.
FASEB J ; 34(5): 6017-6026, 2020 05.
Article in English | MEDLINE | ID: mdl-32306452

ABSTRACT

Human angiotensin-converting enzyme 2 (ACE2) facilitates cellular entry of severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2 as their common receptor. During infection, ACE2-expressing tissues become direct targets, resulting in serious pathological changes and progressive multiple organ failure or even death in severe cases. However, as an essential component of renin-angiotensin system (RAS), ACE2 confers protective effects in physiological circumstance, including maintaining cardiovascular homeostasis, fluid, and electrolyte balance. The absence of protective role of ACE2 leads to dysregulated RAS and thus acute changes under multiple pathological scenarios including SARS. This potentially shared mechanism may also be the molecular explanation for pathogenesis driven by SARS-CoV-2. We reasonably speculate several potential directions of clinical management including host-directed therapies aiming to restore dysregulated RAS caused by ACE2 deficiency. Enriched knowledge of ACE2 learned from SARS and COVID-19 outbreaks can provide, despite their inherent tragedy, informative clues for emerging pandemic preparedness.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/enzymology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/enzymology , Severe Acute Respiratory Syndrome/enzymology , Severe acute respiratory syndrome-related coronavirus/physiology , Virus Internalization , Angiotensin-Converting Enzyme 2 , COVID-19 , Drug Delivery Systems , Humans , Pandemics , Peptidyl-Dipeptidase A/deficiency , SARS-CoV-2
8.
J Biol Chem ; 291(17): 9218-32, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26953343

ABSTRACT

Ebola virus infection can cause severe hemorrhagic fever with a high mortality in humans. The outbreaks of Ebola viruses in 2014 represented the most serious Ebola epidemics in history and greatly threatened public health worldwide. The development of additional effective anti-Ebola therapeutic agents is therefore quite urgent. In this study, via high throughput screening of Food and Drug Administration-approved drugs, we identified that teicoplanin, a glycopeptide antibiotic, potently prevents the entry of Ebola envelope pseudotyped viruses into the cytoplasm. Furthermore, teicoplanin also has an inhibitory effect on transcription- and replication-competent virus-like particles, with an IC50 as low as 330 nm Comparative analysis further demonstrated that teicoplanin is able to block the entry of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) envelope pseudotyped viruses as well. Teicoplanin derivatives such as dalbavancin, oritavancin, and telavancin can also inhibit the entry of Ebola, MERS, and SARS viruses. Mechanistic studies showed that teicoplanin blocks Ebola virus entry by specifically inhibiting the activity of cathepsin L, opening a novel avenue for the development of additional glycopeptides as potential inhibitors of cathepsin L-dependent viruses. Notably, given that teicoplanin has routinely been used in the clinic with low toxicity, our work provides a promising prospect for the prophylaxis and treatment of Ebola, MERS, and SARS virus infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cathepsin L/antagonists & inhibitors , Ebolavirus/metabolism , Endosomes/enzymology , Lysosomes/enzymology , Middle East Respiratory Syndrome Coronavirus/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Teicoplanin/pharmacokinetics , Virus Internalization/drug effects , Cathepsin L/metabolism , Ebolavirus/genetics , Endosomes/genetics , Endosomes/virology , HeLa Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/enzymology , Hemorrhagic Fever, Ebola/epidemiology , Humans , Lysosomes/genetics , Lysosomes/virology , Middle East Respiratory Syndrome Coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/epidemiology
9.
J Virol ; 88(2): 1293-307, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227843

ABSTRACT

The type II transmembrane serine proteases TMPRSS2 and HAT can cleave and activate the spike protein (S) of the severe acute respiratory syndrome coronavirus (SARS-CoV) for membrane fusion. In addition, these proteases cleave the viral receptor, the carboxypeptidase angiotensin-converting enzyme 2 (ACE2), and it was proposed that ACE2 cleavage augments viral infectivity. However, no mechanistic insights into this process were obtained and the relevance of ACE2 cleavage for SARS-CoV S protein (SARS-S) activation has not been determined. Here, we show that arginine and lysine residues within ACE2 amino acids 697 to 716 are essential for cleavage by TMPRSS2 and HAT and that ACE2 processing is required for augmentation of SARS-S-driven entry by these proteases. In contrast, ACE2 cleavage was dispensable for activation of the viral S protein. Expression of TMPRSS2 increased cellular uptake of soluble SARS-S, suggesting that protease-dependent augmentation of viral entry might be due to increased uptake of virions into target cells. Finally, TMPRSS2 was found to compete with the metalloprotease ADAM17 for ACE2 processing, but only cleavage by TMPRSS2 resulted in augmented SARS-S-driven entry. Collectively, our results in conjunction with those of previous studies indicate that TMPRSS2 and potentially related proteases promote SARS-CoV entry by two separate mechanisms: ACE2 cleavage, which might promote viral uptake, and SARS-S cleavage, which activates the S protein for membrane fusion. These observations have interesting implications for the development of novel therapeutics. In addition, they should spur efforts to determine whether receptor cleavage promotes entry of other coronaviruses, which use peptidases as entry receptors.


Subject(s)
ADAM Proteins/metabolism , Peptidyl-Dipeptidase A/metabolism , Serine Endopeptidases/metabolism , Severe Acute Respiratory Syndrome/enzymology , Severe acute respiratory syndrome-related coronavirus/physiology , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization , ADAM Proteins/genetics , ADAM17 Protein , Amino Acid Motifs , Angiotensin-Converting Enzyme 2 , Cell Line , Humans , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/genetics , Protein Processing, Post-Translational , Proteolysis , Receptors, Virus/genetics , Receptors, Virus/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Serine Endopeptidases/genetics , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/metabolism , Severe Acute Respiratory Syndrome/virology , Spike Glycoprotein, Coronavirus/genetics
10.
Bioorg Med Chem ; 20(19): 5928-35, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22884354

ABSTRACT

In the search for anti-SARS-CoV, tanshinones derived from Salvia miltiorrhiza were found to be specific and selective inhibitors for the SARS-CoV 3CL(pro) and PL(pro), viral cysteine proteases. A literature search for studies involving the seven isolated tanshinone hits showed that at present, none have been identified as coronaviral protease inhibitors. We have identified that all of the isolated tanshinones are good inhibitors of both cysteine proteases. However, their activity was slightly affected by subtle changes in structure and targeting enzymes. All isolated compounds (1-7) act as time dependent inhibitors of PL(pro), but no improved inhibition was observed following preincubation with the 3CL(pro). In a detail kinetic mechanism study, all of the tanshinones except rosmariquinone (7) were identified as noncompetitive enzyme isomerization inhibitors. However, rosmariquinone (7) showed a different kinetic mechanism through mixed-type simple reversible slow-binding inhibition. Furthermore, tanshinone I (5) exhibited the most potent nanomolar level inhibitory activity toward deubiquitinating (IC(50)=0.7 µM). Additionally, the inhibition is selective because these compounds do not exert significant inhibitory effects against other proteases including chymotrysin, papain, and HIV protease. These findings provide potential inhibitors for SARS-CoV viral infection and replication.


Subject(s)
Abietanes/chemistry , Abietanes/pharmacology , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Salvia miltiorrhiza/chemistry , Severe acute respiratory syndrome-related coronavirus/enzymology , Abietanes/isolation & purification , Cysteine Proteinase Inhibitors/isolation & purification , Humans , Kinetics , Protein Binding , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/enzymology , Ubiquitination/drug effects
11.
PLoS One ; 7(4): e35876, 2012.
Article in English | MEDLINE | ID: mdl-22558251

ABSTRACT

The type II transmembrane serine proteases TMPRSS2 and HAT activate influenza viruses and the SARS-coronavirus (TMPRSS2) in cell culture and may play an important role in viral spread and pathogenesis in the infected host. However, it is at present largely unclear to what extent these proteases are expressed in viral target cells in human tissues. Here, we show that both HAT and TMPRSS2 are coexpressed with 2,6-linked sialic acids, the major receptor determinant of human influenza viruses, throughout the human respiratory tract. Similarly, coexpression of ACE2, the SARS-coronavirus receptor, and TMPRSS2 was frequently found in the upper and lower aerodigestive tract, with the exception of the vocal folds, epiglottis and trachea. Finally, activation of influenza virus was conserved between human, avian and porcine TMPRSS2, suggesting that this protease might activate influenza virus in reservoir-, intermediate- and human hosts. In sum, our results show that TMPRSS2 and HAT are expressed by important influenza and SARS-coronavirus target cells and could thus support viral spread in the human host.


Subject(s)
Disease Reservoirs/veterinary , Gastrointestinal Tract/enzymology , Influenza, Human/enzymology , Respiratory System/enzymology , Serine Endopeptidases/genetics , Severe Acute Respiratory Syndrome/enzymology , Angiotensin-Converting Enzyme 2 , Animals , Birds , Cell Line , Disease Reservoirs/virology , Enzyme Activation , Gastrointestinal Tract/virology , Gene Expression , Humans , Influenza, Human/genetics , Influenza, Human/transmission , Influenza, Human/virology , Orthomyxoviridae/physiology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Respiratory System/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Serine Endopeptidases/metabolism , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/transmission , Severe Acute Respiratory Syndrome/virology , Sialic Acids/metabolism , Swine
12.
Curr Comput Aided Drug Des ; 6(1): 1-23, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20370692

ABSTRACT

Cysteine proteases are implicated in a variety of human physiological processes and also form an essential component of the life cycle of a number of pathogenic protozoa and viruses. The present review highlights the drug design approaches utilized to understand the mechanism of inhibition and discovery of inhibitors against protozoal cysteine protease, falcipain (a cysteine protease of P. falciparum which causes malaria), and viral cysteine protease, SARS-CoV M(pro) (a cysteine protease of severe acute respiratory syndrome corona virus). The article describes rational approaches for the design of inhibitors and focuses on a variety of structure as well as ligand-based modeling strategies adopted for the discovery of the inhibitors. Also, the key features of ligand recognition against these targets are accentuated. Although no apparent similarities exist between viral and protozoal cysteine proteases discussed here, the goal is to provide examples of rational drug design approaches adopted to design inhibitors against these proteases. The current review would be of interest to scientists engaged in the development of drug design strategies to target the cysteine proteases present in mammals and other lower order organisms.


Subject(s)
Computational Biology/methods , Cysteine Proteinase Inhibitors/chemistry , Drug Discovery/methods , Malaria/drug therapy , Malaria/enzymology , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/enzymology , Animals , Cysteine Proteinase Inhibitors/therapeutic use , Humans
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(9): 777-9, 2009 Sep.
Article in Chinese | MEDLINE | ID: mdl-19737459

ABSTRACT

AIM: Severe acute respiratory syndrome coronavirus (SARS-CoV) is the etiological agent of SARS, an emerging disease characterized by atypical pneumonia. Using a yeast two-hybrid screen with the nucleocapsid (N)protein of SARS-CoV as a bait, the N protein was found to interact with MAP19, a non-enzymatic protein of MASP(mannan-associated serine protease). The interaction between SARS-CoV N and MAP19 would be further tested in cells in this article. METHODS: The interaction between SARS-CoV N and MAP19 was demonstrated by immuno- coprecipitation, and the amount of MAP19 influenced by SARS-CoV N was investgated by Western blot. RESULTS: The interaction between SARS-CoV N and MAP19 was already demonstrated by immuno-coprecipitation. SARS-CoV N greatly increased the amount of MAP19. CONCLUSION: SARS-CoV N can bind with MAP19 in cells. Our study may be conductive to further research into the molecular mechanism of action between SARS-CoV N and MAP19.


Subject(s)
Mannose-Binding Protein-Associated Serine Proteases/metabolism , Nucleocapsid Proteins/metabolism , Severe Acute Respiratory Syndrome/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Cell Line , Coronavirus Nucleocapsid Proteins , Humans , Mannose-Binding Protein-Associated Serine Proteases/genetics , Nucleocapsid Proteins/genetics , Protein Binding , Severe acute respiratory syndrome-related coronavirus/genetics , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/virology
14.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L84-96, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19411314

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is a terminal carboxypeptidase and the receptor for the SARS and NL63 coronaviruses (CoV). Loss of ACE2 function is implicated in severe acute respiratory syndrome (SARS) pathogenesis, but little is known about ACE2 biogenesis and activity in the airways. We report that ACE2 is shed from human airway epithelia, a site of SARS-CoV infection. The regulation of ACE2 release was investigated in polarized human airway epithelia. Constitutive generation of soluble ACE2 was inhibited by DPC 333, implicating a disintegrin and metalloprotease 17 (ADAM17). Phorbol ester, ionomycin, endotoxin, and IL-1beta and TNFalpha acutely induced ACE2 release, further supporting that ADAM17 and ADAM10 regulate ACE2 cleavage. Soluble ACE2 was enzymatically active and partially inhibited virus entry into target cells. We determined that the ACE2 cleavage site resides between amino acid 716 and the putative transmembrane domain starting at amino acid 741. To reveal structural determinants underlying ACE2 release, several mutant and chimeric ACE2 proteins were engineered. Neither the juxtamembrane stalk region, transmembrane domain, nor the cytosolic domain was needed for constitutive ACE2 release. Interestingly, a point mutation in the ACE2 ectodomain, L584A, markedly attenuated shedding. The resultant ACE2-L584A mutant trafficked to the cell membrane and facilitated SARS-CoV entry into target cells, suggesting that the ACE2 ectodomain regulates its release and that residue L584 might be part of a putative sheddase "recognition motif." Thus ACE2 must be cell associated to serve as a CoV receptor and soluble ACE2 might play a role in modifying inflammatory processes at the airway mucosal surface.


Subject(s)
Epithelial Cells/enzymology , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/metabolism , Respiratory System/cytology , Angiotensin-Converting Enzyme 2 , Cell Line , Cell Membrane/metabolism , Cell Polarity , Enzyme Activation , Epithelial Cells/cytology , Humans , Models, Molecular , Mutant Proteins/metabolism , Protein Structure, Tertiary , Severe acute respiratory syndrome-related coronavirus/physiology , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/virology , Solubility , Virus Internalization
15.
J Biol Chem ; 284(12): 7646-55, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19144641

ABSTRACT

Human coxsackievirus (CV) belongs to the picornavirus family, which consists of over 200 medically relevant viruses. In picornavirus, a chymotrypsin-like protease (3C(pro)) is required for viral replication by processing the polyproteins, and thus it is regarded as an antiviral drug target. A 3C-like protease (3CL(pro)) also exists in human coronaviruses (CoV) such as 229E and the one causing severe acute respiratory syndrome (SARS). To combat SARS, we previously had developed peptidomimetic and zinc-coordinating inhibitors of 3CL(pro). As shown in the present study, some of these compounds were also found to be active against 3C(pro) of CV strain B3 (CVB3). Several crystal structures of 3C(pro) from CVB3 and 3CL(pro) from CoV-229E and SARS-CoV in complex with the inhibitors were solved. The zinc-coordinating inhibitor is tetrahedrally coordinated to the His(40)-Cys(147) catalytic dyad of CVB3 3C(pro). The presence of specific binding pockets for the residues of peptidomimetic inhibitors explains the binding specificity. Our results provide a structural basis for inhibitor optimization and development of potential drugs for antiviral therapies.


Subject(s)
Biomimetic Materials/chemistry , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemistry , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/chemistry , Zinc/chemistry , Binding Sites , Biomimetic Materials/therapeutic use , Coronavirus 3C Proteases , Cysteine Proteinase Inhibitors/therapeutic use , Humans , Protein Structure, Tertiary , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/enzymology , Structure-Activity Relationship , Viral Proteins/antagonists & inhibitors
16.
Virology ; 381(1): 89-97, 2008 Nov 10.
Article in English | MEDLINE | ID: mdl-18801550

ABSTRACT

Angiotensin converting enzyme 2 (ACE2) is the receptor that severe acute respiratory syndrome coronavirus (SARS-CoV) utilizes for target cell entry and, therefore, plays an important role in SARS pathogenesis. Since Chinese rhesus (rh) macaques do not usually develop SARS after SARS-CoV infection, it has been suggested that rh-ACE2 probably does not support viral entry efficiently. To determine the role of rh-ACE2 in early lung pathogenesis in vivo, we studied eleven Chinese rhesus monkeys experimentally infected with a pathogenic SARS-CoV(PUMC01) strain. Rh-ACE2 genes were amplified from all animals by reverse transcription polymerase chain reaction, and their function was studied in vitro using a pseudovirus entry assay. Many natural non-synonymous (NS) changes were found in rh-ACE2 genes. Compared to human (hu) ACE2, thirty-eight consensus NS changes were found in rh-ACE2. Since these changes do not interact with the receptor binding domain of SARS-CoV, rh-ACE2 in general is as effective as human homolog in supporting viral entry. Rh-ACE2, however, is more polymorphic than hu-ACE2. Additional sporadic NS substitutions in clone Rh11-7 reduced the level of rh-ACE2 protein expression and did not support viral entry effectively. Further mutagenesis analysis showed that a natural mutation Y217N dramatically alters ACE2 expression and entry efficiency. Moreover, introduction of the Y217N mutation into hu-ACE2 caused the down-regulation of expression and reduced viral entry efficiency. These results indicate that the Y217N mutation plays a role in modulating SARS-CoV infection. Our results provide insights for understanding the role of rh-ACE2 in SARS lung pathogenesis in a non-human primate model.


Subject(s)
Macaca mulatta/virology , Peptidyl-Dipeptidase A/metabolism , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Virus Internalization , Amino Acid Sequence , Angiotensin-Converting Enzyme 2 , Animals , Cell Line , Female , Gene Expression Regulation, Enzymologic , Humans , Lung/enzymology , Lung/pathology , Lung/virology , Male , Molecular Sequence Data , Mutation , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Severe Acute Respiratory Syndrome/pathology
17.
Vet Pathol ; 45(4): 551-62, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18587105

ABSTRACT

The pathology of severe acute respiratory syndrome-coronavirus (SARS-CoV) infection in cats and ferrets is poorly described, and the distribution of angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV, in the respiratory tracts of these species is unknown. We observed SARS-CoV antigen expression and lesions in the respiratory tracts of 4 cats and 4 ferrets at 4 days postinoculation and ACE2 expression in the respiratory tracts of 3 cats and 3 ferrets without infection. All infected cats and ferrets had diffuse alveolar damage associated with SARS-CoV antigen expression. A novel SARS-CoV-associated lesion was tracheo-bronchoadenitis in cats. SARS-CoV antigen expression occurred mainly in type I and II pneumocytes and serous cells of tracheo-bronchial submucosal glands of cats and in type II pneumocytes of ferrets. ACE2 expression occurred mainly in type I and II pneumocytes, tracheo-bronchial goblet cells, serous epithelial cells of tracheo-bronchial submucosal glands in cats, and type II pneumocytes and serous epithelial cells of tracheo-bronchial submucosal glands in ferrets. In conclusion, the pathology of SARS-CoV infection in cats and ferrets resembles that in humans except that syncytia and hyaline membranes were not observed. The identification of tracheo-bronchoadenitis in cats has potential implications for SARS pathogenesis and SARS-CoV excretion. Finally, these results show the importance of ACE2 expression for SARS-CoV infection in vivo: whereas ACE2 expression in type I and II pneumocytes in cats corresponded to SARS-CoV antigen expression in both cell types, expression of both ACE2 and SARS-CoV antigen in ferrets was limited mainly to type II pneumocytes.


Subject(s)
Cats/virology , Disease Models, Animal , Ferrets/virology , Respiratory Tract Infections/virology , Severe Acute Respiratory Syndrome/virology , Severe acute respiratory syndrome-related coronavirus/growth & development , Angiotensin-Converting Enzyme 2 , Animals , Antigens, Viral/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Peptidyl-Dipeptidase A/metabolism , Respiratory Tract Infections/enzymology , Respiratory Tract Infections/pathology , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/pathology , Specific Pathogen-Free Organisms
18.
Cell Mol Life Sci ; 64(15): 2006-12, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17558469

ABSTRACT

Angiotensin-converting enzyme (ACE) and ACE2 are highly homologous metalloproteases that provide essential catalytic functions in the renin-angiotensin system (RAS). Angiotensin II is one key effector peptide of the RAS, inducing vasoconstriction and exerting multiple biological functions. ACE cleaves angiotensin I to generate angiotensin II, whereas ACE2 reduces angiotensin II levels. Accumulating evidence has demonstrated a physiological and pathological role of ACE2 in the cardiovascular systems. Intriguingly, the SARS coronavirus, the cause of severe acute respiratory syndrome (SARS), utilizes ACE2 as an essential receptor for cell fusion and in vivo infections. Moreover, recent studies have demonstrated that ACE2 protects murine lungs from acute lung injury as well as SARS-Spike protein-mediated lung injury, suggesting a dual role of ACE2 in SARS infections and protection from ARDS.


Subject(s)
Peptidyl-Dipeptidase A/metabolism , Respiratory Distress Syndrome/enzymology , Angiotensin-Converting Enzyme 2 , Animals , Humans , Mice , Mice, Knockout , Models, Biological , Peptidyl-Dipeptidase A/deficiency , Peptidyl-Dipeptidase A/genetics , Pulmonary Edema/enzymology , Renin-Angiotensin System/physiology , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/prevention & control , Severe Acute Respiratory Syndrome/enzymology
19.
Biochemistry ; 45(50): 14908-16, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17154528

ABSTRACT

Severe acute respiratory syndrome (SARS) is an infectious disease caused by the human coronavirus, SARS-CoV. The main viral protease, SARS 3CLpro, is a validated target for the development of antiviral therapies. Since the enzyme is a homodimer and the individual monomers are inactive, two approaches are being used to develop inhibitors: enzyme activity inhibitors that target the active site and dimerization inhibitors. Dimerization inhibitors are usually targeted to the dimerization interface and need to compete with the attractive forces between subunits to be effective. In this paper, we show that the dimerization of SARS 3CLpro is also under allosteric control and that additional and energetically more favorable target sites away from the dimerization interface may also lead to subunit dissociation. We previously identified a cluster of conserved serine residues (Ser139, Ser144, and Ser147) located adjacent to the active site of 3CLpro that could effectively be targeted to inactivate the protease [Bacha, U et al. (2004) Biochemistry 43, 4906-4912]. Mutation of any of these serine residues to alanine had a debilitating effect on the catalytic activity of 3CLpro. In particular, the mutation of Ser147, which does not make any contact with the opposing subunit and is located approximately 9 A away from the dimer interface, totally inhibited dimerization and resulted in a complete loss of enzymatic activity. The finding that residues away from the dimer interface are able to control dimerization defines alternative targets for the design of dimerization inhibitors.


Subject(s)
Cysteine Endopeptidases/chemistry , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/chemistry , Allosteric Site/genetics , Amino Acid Substitution , Binding Sites/genetics , Coronavirus 3C Proteases , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Dimerization , Humans , Protein Binding/genetics , Protein Structure, Quaternary/genetics , Protein Structure, Tertiary/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Serine/chemistry , Serine/genetics , Serine/metabolism , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
20.
J Mol Med (Berl) ; 84(10): 814-20, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16988814

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2), a second angiotensin-converting enzyme (ACE), regulates the renin-angiotensin system by counterbalancing ACE activity. Accumulating evidence in recent years has demonstrated a physiological and pathological role of ACE2 in the cardiovascular systems. Recently, it has been shown that severe acute respiratory syndrome (SARS) coronavirus, the cause of SARS, utilizes ACE2 as an essential receptor for cell fusion and in vivo infections in mice. Intriguingly, ACE2 acts as a protective factor in various experimental models of acute lung failure and, therefore, acts not only as a key determinant for SARS virus entry into cells but also contributes to SARS pathogenesis. Here we review the role of ACE2 in disease pathogenesis, including lung diseases and cardiovascular diseases.


Subject(s)
Lung Diseases/enzymology , Peptidyl-Dipeptidase A/metabolism , Severe Acute Respiratory Syndrome/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/therapy , Genetic Therapy/methods , Humans , Lung Diseases/metabolism , Lung Diseases/therapy , Mice , Models, Biological , Peptidyl-Dipeptidase A/genetics , Severe acute respiratory syndrome-related coronavirus/growth & development , Severe Acute Respiratory Syndrome/enzymology , Severe Acute Respiratory Syndrome/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...