Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: covidwho-2243576

ABSTRACT

Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.


Subject(s)
Cardiovascular Diseases , Kidney Diseases , Receptors, sigma , Humans , Cardiomegaly , COVID-19/complications , Heart Failure/complications , Ligands , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/genetics , Receptors, sigma/metabolism , Signal Transduction/physiology , Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Kidney Diseases/complications , Kidney Diseases/genetics , Kidney Diseases/metabolism
2.
Salus ; 25(3):25-31, 2021.
Article in Spanish | EMBASE | ID: covidwho-2207151

ABSTRACT

Background: SARS-CoV-2, the etiological agent responsible for COVID-19, is an RNA virus belonging to the Coronaviridae family. During the virus replication, viral components interact with the cellular machinery, inducing alterations in cell physiology, which contributes to viral pathogenesis. Method(s): A bibliographical research about cellular stress and SARS-CoV-2 was performed at NCBI/Pubmed. Result(s): In response to the infection, signaling pathways are activated in the host cell, the goal of these pathways being to restore homeostasis. If homeostasis is not recovered, the signaling leads to cell death activation. Among the best-characterized signaling pathways, the cellular stress pathways such as oxidative stress, UPR, and autophagy stand out, which are evolutionarily conserved and are also interconnected with each other. There is strong theoretical and experimental evidence of various interactions of some components of these pathways with different viral proteins of coronavirus, and some studies with SARS-CoV-2 have already been performed. In this review, we highlight some of the cellular pathways-virus characterized to date. Conclusion(s): The cellular pathways and their relationship to viral infections remains unclear. The study of these relationships might constitute an important target for new research and the development of antiviral therapies. Copyright © 2021, Revista Salus. All rights reserved.

3.
Coronavirus Drug Discovery: Druggable Targets and In Silico Update: Volume 3 ; : 23-49, 2022.
Article in English | Scopus | ID: covidwho-2149157

ABSTRACT

The occurrence of SARS-CoV-2 in 2019 is the second coronavirus spread-out after the SARS-CoV, which has pandemic potential. Search for its remedies is dependent on integrative knowledge of cell signaling pathways, which is under clinical scrutiny. The major cascades triggered by coronavirus entry include Renin-Angiotensin System, MAPK, NF-κB, JAK/STAT which are involved with innate immunity. Some other modes are, unfolded protein response signaling and inflammasome mediated apoptosis activation. Virulence factors of the SARS-CoVs like spike, envelope, nonstructural proteins etc., interfere with some of these viral defense pathways. Therapeutically, the viral intrusion, multiplication, as well as tissue-injurious cytokine overreactions are targeted by pathway-specific drugs. Viral entry blockers, p38 MAPK inhibitors, cytokine regulators, JAK inhibitors, and anti-inflammatory drugs are either being repurposed or innovated with scopes for futuristic modeling. This chapter is aimed to elucidate the pathological signaling network behind Severe Acute Respiratory Syndrome, for evaluation of existing and postulated drug targets. © 2022 Elsevier Inc. All rights reserved.

4.
J Migr Health ; 6: 100137, 2022.
Article in English | MEDLINE | ID: covidwho-2041957

ABSTRACT

Access to birth registration among the refugees, migrants, and undocumented or stateless individuals in Sabah and Peninsular Malaysia remains hindered largely due to their lack of legal status. This study identifies the barriers to birth registration faced by these communities, including during the COVID-19 pandemic, and explores the extent to which digital technologies may overcome or amplify these barriers. Findings are reported from a review of literature, websites, and media articles and semi-structured interviews with community-based organisations and community leaders representing the communities. The themes for the questions were structured based on Plan International's (2015) Step-by-step Guide for Identifying and Addressing the Risks to Children in Digitised birth registration systems. We identified that the digitalisation of birth registration poses more risks of exclusion than benefits to the marginalised communities without a secure and inclusive operating environment. Subject to an inequality assessment to evaluate and address the existing inequalities, a hybrid system that factors in the role of citizen facilitation hubs would be ideal for ensuring no one gets "left behind".

5.
Pathogens ; 11(8)2022 Aug 19.
Article in English | MEDLINE | ID: covidwho-1997737

ABSTRACT

Swine coronaviruses include the following six members, namely porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), porcine delta coronavirus (PDCoV), swine acute diarrhea syndrome coronavirus (SADS-CoV), porcine hemagglutinating encephalomyelitis virus (PHEV), and porcine respiratory coronavirus (PRCV). Clinically, PEDV, TGEV, PDCoV, and SADS-CoV cause enteritis, whereas PHEV induces encephalomyelitis, and PRCV causes respiratory disease. Years of studies reveal that swine coronaviruses replicate in the cellular cytoplasm exerting a wide variety of effects on cells. Some of these effects are particularly pertinent to cell pathology, including endoplasmic reticulum (ER) stress, unfolded protein response (UPR), autophagy, and apoptosis. In addition, swine coronaviruses are able to induce cellular changes, such as cytoskeletal rearrangement, alterations of junctional complexes, and epithelial-mesenchymal transition (EMT), that render enterocytes unable to absorb nutrients normally, resulting in the loss of water, ions, and protein into the intestinal lumen. This review aims to describe the cellular changes in swine coronavirus-infected cells and to aid in understanding the pathogenesis of swine coronavirus infections. This review also explores how the virus exerted subcellular and molecular changes culminating in the clinical and pathological findings observed in the field.

6.
Front Cell Neurosci ; 16: 905218, 2022.
Article in English | MEDLINE | ID: covidwho-1974665

ABSTRACT

We are living in a terrifying pandemic caused by Sars-CoV-2, in which patients with diabetes mellitus have, from the beginning, been identified as having a high risk of hospitalization and mortality. This viral disease is not limited to the respiratory system, but also affects, among other organs, the central nervous system. Furthermore, we already know that individuals with diabetes mellitus exhibit signs of astrocyte dysfunction and are more likely to develop cognitive deficits and even dementia. It is now being realized that COVID-19 incurs long-term effects and that those infected can develop several neurological and psychiatric manifestations. As this virus seriously compromises cell metabolism by triggering several mechanisms leading to the unfolded protein response (UPR), which involves endoplasmic reticulum Ca2+ depletion, we review here the basis involved in this response that are intimately associated with the development of neurodegenerative diseases. The discussion aims to highlight two aspects-the role of calcium-binding proteins and the role of astrocytes, glial cells that integrate energy metabolism with neurotransmission and with neuroinflammation. Among the proteins discussed are calpain, calcineurin, and sorcin. These proteins are emphasized as markers of the UPR and are potential therapeutic targets. Finally, we discuss the role of drugs widely prescribed to patients with diabetes mellitus, such as statins, metformin, and calcium channel blockers. The review assesses potential neuroprotection mechanisms, focusing on the UPR and the restoration of reticular Ca2+ homeostasis, based on both clinical and experimental data.

7.
Molecules ; 27(14)2022 Jul 06.
Article in English | MEDLINE | ID: covidwho-1917640

ABSTRACT

Different pathological conditions, including viral infections and cancer, can have a massive impact on the endoplasmic reticulum (ER), causing severe damage to the cell and exacerbating the disease. In particular, coronavirus infections, including SARS coronavirus-2 (SARS-CoV-2), responsible for COVID-19, cause ER stress as a consequence of the enormous amounts of viral glycoproteins synthesized, the perturbation of ER homeostasis and the modification of ER membranes. Therefore, ER has a central role in the viral life cycle, thus representing one of the Achilles' heels on which to focus therapeutic intervention. On the other hand, prolonged ER stress has been demonstrated to promote many pro-tumoral attributes in cancer cells, having a key role in tumor growth, metastasis and response to therapies. In this report, adopting a repurposing approach of approved drugs, we identified the antiplatelet agent ticlopidine as an interferent of the unfolded protein response (UPR) via sigma receptors (SRs) modulation. The promising results obtained suggest the potential use of ticlopidine to counteract ER stress induced by viral infections, such as COVID-19, and cancer.


Subject(s)
COVID-19 Drug Treatment , Neoplasms , Drug Repositioning , Endoplasmic Reticulum Stress , Humans , Neoplasms/pathology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , SARS-CoV-2 , Ticlopidine/pharmacology , Unfolded Protein Response
8.
Vet Microbiol ; 271: 109494, 2022 Aug.
Article in English | MEDLINE | ID: covidwho-1886124

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging enteropathogenic coronavirus that has the potential for cross-species infection. Many viruses have been reported to induce endoplasmic reticulum stress (ERS) and activate the unfolded protein response (UPR). To date, little is known about whether and, if so, how the UPR is activated by PDCoV infection. Here, we investigated the activation state of UPR pathways and their effects on viral replication during PDCoV infection. We found that PDCoV infection induced ERS and activated all three known UPR pathways (inositol-requiring enzyme 1 [IRE1], activating transcription factor 6 [ATF6], and PKR-like ER kinase [PERK]), as demonstrated by IRE1-mediated XBP1 mRNA cleavage and increased mRNA expression of XBP1s, ATF4, CHOP, GADD34, GRP78, and GRP94, as well as phosphorylated eIF2α expression. Through pharmacologic treatment, RNA interference, and overexpression experiments, we confirmed the negative role of the PERK-eIF2α pathway and the positive regulatory role of the ATF6 pathway, but found no obvious effect of IRE1 pathway, on PDCoV replication. Taken together, our results characterize, for the first time, the state of the ERS response during PDCoV infection and identify the PERK and ATF6 pathways as potential antiviral targets.


Subject(s)
Protein Serine-Threonine Kinases , Unfolded Protein Response , Animals , Deltacoronavirus , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Swine , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
9.
Adv Exp Med Biol ; 1352: 125-147, 2021.
Article in English | MEDLINE | ID: covidwho-1669700

ABSTRACT

INTRODUCTION: The recent outbreak of coronavirus infection by SARS-CoV-2 that started from the Wuhan Province of China in 2019 has spread to most parts of the world infecting millions of people. Although the case fatality rate of SARS-CoV-2 infection is less than the previous epidemics by other closely related coronaviruses, due to its high infectivity, the total number of SARS-CoV-2 infection-associated disease, called Covid-19, is a matter of global concern. Despite drastic preventive measures, the number of Covid-19 cases are steadily increasing, and the future course of this pandemic is highly unpredictable. The most concerning fact about Covid-19 is the absence of specific and effective preventive or therapeutic agents against the disease. Finding an immediate intervention against Covid-19 is the need of the hour. In this chapter, we have discussed the role of different branches of the cellular proteostasis network, represented by Hsp70-Hsp40 chaperone system, Ubiquitin-Proteasome System (UPS), autophagy, and endoplasmic reticulum-Unfolded Protein Response (ER-UPR) pathway in the pathogenesis of coronavirus infections and in the host antiviral defense mechanisms. RESULTS: Based on scientific literature, we present that pharmacological manipulation of proteostasis network can alter the fate of coronavirus infections and may help to prevent the resulting pathologies like Covid-19.


Subject(s)
COVID-19 , Humans , Pandemics , Proteostasis , SARS-CoV-2 , Unfolded Protein Response
10.
Viruses ; 13(12)2021 12 09.
Article in English | MEDLINE | ID: covidwho-1572656

ABSTRACT

In the past year and a half, SARS-CoV-2 has caused 240 million confirmed cases and 5 million deaths worldwide. Autophagy is a conserved process that either promotes or inhibits viral infections. Although coronaviruses are known to utilize the transport of autophagy-dependent vesicles for the viral life cycle, the underlying autophagy-inducing mechanisms remain largely unexplored. Using several autophagy-deficient cell lines and autophagy inhibitors, we demonstrated that SARS-CoV-2 ORF3a was able to induce incomplete autophagy in a FIP200/Beclin-1-dependent manner. Moreover, ORF3a was involved in the induction of the UPR (unfolded protein response), while the IRE1 and ATF6 pathways, but not the PERK pathway, were responsible for mediating the ORF3a-induced autophagy. These results identify the role of the UPR pathway in the ORF3a-induced classical autophagy process, which may provide us with a better understanding of SARS-CoV-2 and suggest new therapeutic modalities in the treatment of COVID-19.


Subject(s)
Autophagy , SARS-CoV-2/metabolism , Unfolded Protein Response , Viroporin Proteins/metabolism , Animals , Autophagy/genetics , Autophagy-Related Proteins/genetics , Beclin-1/genetics , Cell Line , Humans , Signal Transduction
11.
Br Med Bull ; 137(1): 13-27, 2021 03 25.
Article in English | MEDLINE | ID: covidwho-1054267

ABSTRACT

BACKGROUND: Many drugs approved for other indications can control the growth of tumor cells and limit adverse events (AE). DATA SOURCES: Literature searches with keywords 'repurposing and cancer' books, websites: https://clinicaltrials.gov/, for drug structures: https://pubchem.ncbi.nlm.nih.gov/. AREAS OF AGREEMENT: Introducing approved drugs, such as those developed to treat diabetes (Metformin) or inflammation (Thalidomide), identified to have cytostatic activity, can enhance chemotherapy or even replace more cytotoxic drugs. Also, anti-inflammatory compounds, cytokines and inhibitors of proteolysis can be used to control the side effects of chemo- and immuno-therapies or as second-line treatments for tumors resistant to kinase inhibitors (KI). Drugs specifically developed for cancer therapy, such as interferons (IFN), the tyrosine KI abivertinib TKI (tyrosine kinase inhibitor) and interleukin-6 (IL-6) receptor inhibitors, may help control symptoms of Covid-19. AREAS OF CONTROVERSY: Better knowledge of mechanisms of drug activities is essential for repurposing. Chemotherapies induce ER stress and enhance mutation rates and chromosome alterations, leading to resistance that cannot always be related to mutations in the target gene. Metformin, thalidomide and cytokines (IFN, tumor necrosis factor (TNF), interleukin-2 (IL-2) and others) have pleiomorphic activities, some of which can enhance tumorigenesis. The small and fragile patient pools available for clinical trials can cloud the data on the usefulness of cotreatments. GROWING POINTS: Better understanding of drug metabolism and mechanisms should aid in repurposing drugs for primary, adjuvant and adjunct treatments. AREAS TIMELY FOR DEVELOPING RESEARCH: Optimizing drug combinations, reducing cytotoxicity of chemotherapeutics and controlling associated inflammation.


Subject(s)
COVID-19 Drug Treatment , Drug Repositioning , Neoplasms/drug therapy , Humans
12.
Brief Funct Genomics ; 20(1): 28-41, 2021 03 02.
Article in English | MEDLINE | ID: covidwho-1045889

ABSTRACT

The human genome has an almost equal distribution of unique and transposable genetic elements. Although at the transcriptome level, a relatively higher contribution from transposable elements derived RNA has been reported. This is further highlighted with evidence from pervasive transcription. Of the total RNA, noncoding RNAs (ncRNAs) are significant contributors to the transcriptome pool with sizeable fraction from repetitive elements of the human genome, inclusive of Long Interspersed Nuclear Elements (LINEs) and Short Interspersed Nuclear Elements (SINEs). ncRNAs are increasingly being implicated in diverse functional roles especially during conditions of stress. These stress responses are driven through diverse mediators, inclusive of long and short ncRNAs. ncRNAs such as MALAT1, GAS5, miR-204 and miR-199a-5p have been functionally involved during oxidative stress, endoplasmic reticulum (ER) stress and unfolded protein response (UPR). Also, within SINEs, Alu RNAs derived from primate-specific Alu repeats with ~11% human genome contribution, playing a significant role. Pathogenic diseases, including the recent COVID-19, leads to differential regulation of ncRNAs. Although, limited evidence suggests the need for an inquest into the role of ncRNAs in determining the host response towards pathogen challenge.


Subject(s)
Infections/genetics , RNA, Untranslated/physiology , COVID-19/genetics , COVID-19/virology , Cytokines/physiology , Endoplasmic Reticulum Stress , Host-Pathogen Interactions , Humans , Infections/metabolism , Long Interspersed Nucleotide Elements , Oxidative Stress , RNA, Untranslated/genetics , SARS-CoV-2/isolation & purification , Short Interspersed Nucleotide Elements , Unfolded Protein Response
13.
Am J Physiol Lung Cell Mol Physiol ; 319(3): L444-L455, 2020 09 01.
Article in English | MEDLINE | ID: covidwho-695277

ABSTRACT

Cold viruses have generally been considered fairly innocuous until the appearance of the severe acute respiratory coronavirus 2 (SARS-CoV-2) in 2019, which caused the coronavirus disease 2019 (COVID-19) global pandemic. Two previous viruses foreshadowed that a coronavirus could potentially have devastating consequences in 2002 [severe acute respiratory coronavirus (SARS-CoV)] and in 2012 [Middle East respiratory syndrome coronavirus (MERS-CoV)]. The question that arises is why these viruses are so different from the relatively harmless cold viruses. On the basis of an analysis of the current literature and using bioinformatic approaches, we examined the potential human miRNA interactions with the SARS-CoV-2's genome and compared the miRNA target sites in seven coronavirus genomes that include SARS-CoV-2, MERS-CoV, SARS-CoV, and four nonpathogenic coronaviruses. Here, we discuss the possibility that pathogenic human coronaviruses, including SARS-CoV-2, could modulate host miRNA levels by acting as miRNA sponges to facilitate viral replication and/or to avoid immune responses.


Subject(s)
Betacoronavirus/immunology , Betacoronavirus/isolation & purification , Coronavirus Infections/virology , MicroRNAs/genetics , MicroRNAs/immunology , Pneumonia, Viral/virology , Virus Replication , COVID-19 , Coronavirus Infections/immunology , Humans , Pandemics , Pneumonia, Viral/immunology , SARS-CoV-2
14.
Front Pharmacol ; 11: 1095, 2020.
Article in English | MEDLINE | ID: covidwho-685173

ABSTRACT

Coronavirus disease 2019 (COVID-19), the seventh human coronavirus infectious disease, was first reported in Wuhan, China, in December 2019, followed by its rapid spread globally (251,059 deaths, on May 5, 2020, by Johns Hopkins University). An early clinical report showed that fever, cough, fatigue, sputum production, and myalgia were initial symptoms, with the development of pneumonia as the disease progressed. Increases in the level of serum liver enzymes, D-dimer, cardiac troponin I, and creatinine have been observed in severely ill patients, indicating that multiple organ failure had occurred in these cases. Lymphopenia and an increase in interleukin-6 (IL-6) were also observed. Although COVID-19 patients are administered glucocorticoid therapy to treat the excessive immune response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, the efficacy of this form of therapy is unclear. Viremia is observed in severe cases, suggesting that in addition to type II alveolar epithelial cells, many cell types, such as vascular endothelial cells, cardiomyocytes, renal tubular cells, neuronal cells, and lymphocytes, may be damaged. The improvement of survival rates requires elucidation of the mechanism by which cellular damage occurs during viral infection. Cellular therapy, along with organ support systems such as oxygen therapy, artificial ventilation, extra corporeal membrane oxygenation and dialysis, as well as antiviral therapy, are required. Viral replication in infected host cells may perturb protein folding in the endoplasmic reticulum (ER), causing ER stress. Although an adaptive cellular response, i.e. the unfolded protein response, can compensate for the misfolded protein burden to some extent, continued viral proliferation may induce inflammation and cell death. Therefore, we propose that proteostasis dysfunction may cause conformational disorders in COVID-19. The application of pharmacological chaperone therapy to treat COVID-19 patients is additionally discussed.

15.
Int J Mol Sci ; 21(10)2020 May 20.
Article in English | MEDLINE | ID: covidwho-324354

ABSTRACT

The COVID-19 global pandemic is caused by SARS-CoV-2, and represents an urgent medical and social issue. Unfortunately, there is still not a single proven effective drug available, and therefore, current therapeutic guidelines recommend supportive care including oxygen administration and treatment with antibiotics. Recently, patients have been also treated with off-label therapies which comprise antiretrovirals, anti-inflammatory compounds, antiparasitic agents and plasma from convalescent patients, all with controversial results. The ubiquitin-proteasome system (UPS) is important for the maintenance of cellular homeostasis, and plays a pivotal role in viral replication processes. In this review, we discuss several aspects of the UPS and the effects of its inhibition with particular regard to the life cycle of the coronaviruses (CoVs). In fact, proteasome inhibition by various chemical compounds, such as MG132, epoxomycin and bortezomib, may reduce the virus entry into the eucariotic cell, the synthesis of RNA, and the subsequent protein expression necessary for CoVs. Importantly, since UPS inhibitors reduce the cytokine storm associated with various inflammatory conditions, it is reasonable to assume that they might be repurposed for SARS-CoV-2, thus providing an additional tool to counteract both virus replication as well as its most deleterious consequences triggered by abnormal immunological response.


Subject(s)
Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Proteasome Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Betacoronavirus/drug effects , Betacoronavirus/isolation & purification , COVID-19 , Coronavirus Infections/epidemiology , Endoplasmic Reticulum Stress/drug effects , Humans , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Pandemics , Pneumonia, Viral/epidemiology , Proteasome Inhibitors/pharmacology , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL