Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Microb Risk Anal ; 24: 100263, 2023 Aug.
Article in English | MEDLINE | ID: covidwho-2325617

ABSTRACT

From the perspectives of molecular biology, genetics and biothermodynamics, SARS-CoV-2 is the among the best characterized viruses. Research on SARS-CoV-2 has shed a new light onto driving forces and molecular mechanisms of viral evolution. This paper reports results on empirical formulas, biosynthesis reactions and thermodynamic properties of biosynthesis (multiplication) for the Zeta P.2, Eta B.1.525, Theta P.3, Kappa B.1.617.1, Iota B.1.526, Lambda C.37 and Mu B.1.621 variants of SARS-CoV-2. Thermodynamic analysis has shown that the physical driving forces for evolution of SARS-CoV-2 are Gibbs energy of biosynthesis and Gibbs energy of binding. The driving forces have led SARS-CoV-2 through the evolution process from the original Hu-1 to the newest variants in accordance with the expectations of the evolution theory.

2.
Mol Cell Biochem ; 2022 Oct 29.
Article in English | MEDLINE | ID: covidwho-2313046

ABSTRACT

SARS-CoV-2 is a positive-strand RNA virus that infects humans through the nasopharyngeal and oral route causing COVID-19. Scientists left no stone unturned to explore a targetable key player in COVID-19 pathogenesis against which therapeutic interventions can be initiated. This article has attempted to review, coordinate and accumulate the most recent observations in support of the hypothesis predicting the altered state of mitochondria concerning mitochondrial redox homeostasis, inflammatory regulations, morphology, bioenergetics and antiviral signalling in SARS-CoV-2 infection. Mitochondria is extremely susceptible to physiological as well as pathological stimuli, including viral infections. Recent studies suggest that SARS-CoV-2 pathogeneses alter mitochondrial integrity, in turn mitochondria modulate cellular response against the infection. SARS-CoV-2 M protein inhibited mitochondrial antiviral signalling (MAVS) protein aggregation in turn hinders innate antiviral response. Viral open reading frames (ORFs) also play an instrumental role in altering mitochondrial regulation of immune response. Notably, ORF-9b and ORF-6 impair MAVS activation. In aged persons, the NLRP3 inflammasome is over-activated due to impaired mitochondrial function, increased mitochondrial reactive oxygen species (mtROS), and/or circulating free mitochondrial DNA, resulting in a hyper-response of classically activated macrophages. This article also tries to understand how mitochondrial fission-fusion dynamics is affected by the virus. This review comprehends the overall mitochondrial attribute in pathogenesis as well as prognosis in patients infected with COVID-19 taking into account pertinent in vitro, pre-clinical and clinical data encompassing subjects with a broad range of severity and morbidity. This endeavour may help in exploring novel non-canonical therapeutic strategies to COVID-19 disease and associated complications.

3.
Int J Mol Sci ; 24(1)2022 Dec 29.
Article in English | MEDLINE | ID: covidwho-2216313

ABSTRACT

The mechanistic interplay between SARS-CoV-2 infection, inflammation, and oxygen homeostasis is not well defined. Here, we show that the hypoxia-inducible factor (HIF-1α) transcriptional pathway is activated, perhaps due to a lack of oxygen or an accumulation of mitochondrial reactive oxygen species (ROS) in the lungs of adult Syrian hamsters infected with SARS-CoV-2. Prominent nuclear localization of HIF-1α and increased expression of HIF-1α target proteins, including glucose transporter 1 (Glut1), lactate dehydrogenase (LDH), and pyruvate dehydrogenase kinase-1 (PDK1), were observed in areas of lung consolidation filled with infiltrating monocytes/macrophages. Upregulation of these HIF-1α target proteins was accompanied by a rise in glycolysis as measured by extracellular acidification rate (ECAR) in lung homogenates. A concomitant reduction in mitochondrial respiration was also observed as indicated by a partial loss of oxygen consumption rates (OCR) in isolated mitochondrial fractions of SARS-CoV-2-infected hamster lungs. Proteomic analysis further revealed specific deficits in the mitochondrial ATP synthase (Atp5a1) within complex V and in the ATP/ADP translocase (Slc25a4). The activation of HIF-1α in inflammatory macrophages may also drive proinflammatory cytokine production and complement activation and oxidative stress in infected lungs. Together, these findings support a role for HIF-1α as a central mediator of the metabolic reprogramming, inflammation, and bioenergetic dysfunction associated with SARS-CoV-2 infection.


Subject(s)
COVID-19 , Hypoxia-Inducible Factor 1, alpha Subunit , Oxidative Stress , Cricetinae , COVID-19/metabolism , Energy Metabolism , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation , Oxygen , Proteomics , SARS-CoV-2
4.
Aging Cell ; 21(11): e13727, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2063328

ABSTRACT

There is still a significant lack of knowledge regarding many aspects of the etiopathology and consequences of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans. For example, the variety of molecular mechanisms mediating this infection, and the long-term consequences of the disease remain poorly understood. It first seemed like the SARS-CoV-2 infection primarily caused a serious respiratory syndrome. However, over the last years, an increasing number of studies also pointed towards the damaging effects of this infection has on the central nervous system (CNS). In fact, evidence suggests a possible disruption of the blood-brain barrier and deleterious effects on the CNS, especially in patients who already suffer from other pathologies, such as neurodegenerative disorders. The molecular mechanisms behind these effects on the CNS could involve the dysregulation of mitochondrial physiology, a well-known early marker of neurodegeneration and a hallmark of aging. Moreover, mitochondria are involved in the activation of the inflammatory response, which has also been broadly described in the CNS in COVID-19. Here, we critically review the current bibliography regarding the presence of neurodegenerative symptoms in COVID-19 patients, with a special emphasis on the mitochondrial mechanisms of these disorders.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Blood-Brain Barrier , Central Nervous System , Mitochondria
5.
Int J Mol Sci ; 22(15)2021 Aug 02.
Article in English | MEDLINE | ID: covidwho-1736939

ABSTRACT

Mitochondria are complex intracellular organelles traditionally identified as the powerhouses of eukaryotic cells due to their central role in bioenergetic metabolism. In recent decades, the growing interest in mitochondria research has revealed that these multifunctional organelles are more than just the cell powerhouses, playing many other key roles as signaling platforms that regulate cell metabolism, proliferation, death and immunological response. As key regulators, mitochondria, when dysfunctional, are involved in the pathogenesis of a wide range of metabolic, neurodegenerative, immune and neoplastic disorders. Far more recently, mitochondria attracted renewed attention from the scientific community for their ability of intercellular translocation that can involve whole mitochondria, mitochondrial genome or other mitochondrial components. The intercellular transport of mitochondria, defined as horizontal mitochondrial transfer, can occur in mammalian cells both in vitro and in vivo, and in physiological and pathological conditions. Mitochondrial transfer can provide an exogenous mitochondrial source, replenishing dysfunctional mitochondria, thereby improving mitochondrial faults or, as in in the case of tumor cells, changing their functional skills and response to chemotherapy. In this review, we will provide an overview of the state of the art of the up-to-date knowledge on intercellular trafficking of mitochondria by discussing its biological relevance, mode and mechanisms underlying the process and its involvement in different pathophysiological contexts, highlighting its therapeutic potential for diseases with mitochondrial dysfunction primarily involved in their pathogenesis.


Subject(s)
Metabolic Diseases/physiopathology , Mitochondria/physiology , Mitochondrial Dynamics , Neoplasms/physiopathology , Neurodegenerative Diseases/physiopathology , Animals , Humans , Metabolic Diseases/therapy , Neoplasms/therapy
6.
Int J Mol Sci ; 22(16)2021 Aug 22.
Article in English | MEDLINE | ID: covidwho-1367850

ABSTRACT

SARS-CoV-2 primarily infects epithelial airway cells that express the host entry receptor angiotensin-converting enzyme 2 (ACE2), which binds to the S1 spike protein on the surface of the virus. To delineate the impact of S1 spike protein interaction with the ACE2 receptor, we incubated the S1 spike protein with human pulmonary arterial endothelial cells (HPAEC). HPAEC treatment with the S1 spike protein caused disruption of endothelial barrier function, increased levels of numerous inflammatory molecules (VCAM-1, ICAM-1, IL-1ß, CCL5, CXCL10), elevated mitochondrial reactive oxygen species (ROS), and a mild rise in glycolytic reserve capacity. Because low oxygen tension (hypoxia) is associated with severe cases of COVID-19, we also evaluated treatment with hemoglobin (HbA) as a potential countermeasure in hypoxic and normal oxygen environments in analyses with the S1 spike protein. We found hypoxia downregulated the expression of the ACE2 receptor and increased the critical oxygen homeostatic signaling protein, hypoxia-inducible factor (HIF-1α); however, treatment of the cells with HbA yielded no apparent change in the levels of ACE2 or HIF-1α. Use of quantitative proteomics revealed that S1 spike protein-treated cells have few differentially regulated proteins in hypoxic conditions, consistent with the finding that ACE2 serves as the host viral receptor and is reduced in hypoxia. However, in normoxic conditions, we found perturbed abundance of proteins in signaling pathways related to lysosomes, extracellular matrix receptor interaction, focal adhesion, and pyrimidine metabolism. We conclude that the spike protein alone without the rest of the viral components is sufficient to elicit cell signaling in HPAEC, and that treatment with HbA failed to reverse the vast majority of these spike protein-induced changes.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/pathology , Endothelial Cells/metabolism , Hemoglobins/metabolism , Spike Glycoprotein, Coronavirus/metabolism , COVID-19/virology , Cell Hypoxia , Cell Survival , Cells, Cultured , Endothelial Cells/virology , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Humans , Protein Subunits/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/pathology , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity
7.
Cureus ; 13(7): e16277, 2021 Jul.
Article in English | MEDLINE | ID: covidwho-1332354

ABSTRACT

BACKGROUND AND AIM: The aim of this study is to assess if people who chose to receive the preventive Natural Bioenergetics (NB) COVID-19 treatment would experience safely a strong reduction in frequency and severity of COVID-19 major symptoms (fever, cough, and shortness of breath) compared to the general population. Experimental procedure: The preventive NB COVID-19 treatment is a double acupuncture meridian-based procedure that primes the immune system using acupuncture points and specific substances and sounds on precise body locations. Four hundred and thirty-nine people from seven countries (Canada, USA, Mexico, UK, France, Israel, and Belgium) voluntarily received the non-invasive preventive NB treatment. Data used for this study have been gathered between April 2020 and December 2020. The severity of cases experienced by the general population was statistically compared with those of the 42 infected people of this study. RESULTS AND CONCLUSION: Our analysis suggests the population who chose to receive the preventive NB COVID-19 treatment experienced a strong reduction in frequency and severity of the three major symptoms of COVID-19 (p<0.01) compared to the general population. Nobody in that population needed hospitalization, including the elderly, which can be interpreted as a very significant clinical improvement. Most people did not report any side effects. Only small side effects were reported.

8.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L358-L376, 2021 08 01.
Article in English | MEDLINE | ID: covidwho-1280497

ABSTRACT

Capillary endothelial cells possess a specialized metabolism necessary to adapt to the unique alveolar-capillary environment. Here, we highlight how endothelial metabolism preserves the integrity of the pulmonary circulation by controlling vascular permeability, defending against oxidative stress, facilitating rapid migration and angiogenesis in response to injury, and regulating the epigenetic landscape of endothelial cells. Recent reports on single-cell RNA-sequencing reveal subpopulations of pulmonary capillary endothelial cells with distinctive reparative capacities, which potentially offer new insight into their metabolic signature. Lastly, we discuss broad implications of pulmonary vascular metabolism on acute respiratory distress syndrome, touching on emerging findings of endotheliitis in coronavirus disease 2019 (COVID-19) lungs.


Subject(s)
COVID-19/complications , Endothelium, Vascular/metabolism , Neovascularization, Pathologic/pathology , Pulmonary Circulation , Respiratory Distress Syndrome/epidemiology , SARS-CoV-2/isolation & purification , COVID-19/transmission , COVID-19/virology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Humans , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/virology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology
9.
J Photochem Photobiol B ; 213: 112083, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-1023667

ABSTRACT

In developing an effective clinical tool against COVID-19, we need to consider why SARS-CoV-2 infections develop along remarkably different trajectories: from completely asymptomatic to a severe course of disease. In this paper we hypothesize that the progressive exhaustion and loss of lymphocytes associated with severe stages of COVID-19 result from an intracellular energy deficit in an organism which has already been depleted by preexisting chronic diseases, acute psychological stress and the aging process. A bioenergetics view of COVID-19 immunopathology opens a new biophysical opportunity to enhance impaired immune function via proposed pathways of photomagnetic catalysis of ATP synthesis, regenerative photobiomodulation and the ultrasonic acceleration of cell restructuring. Moreover, we suggest that a coherent application of multiple biophysical radiances (coMra) may synergistically enhance energy-matter-information kinetics of basal self-regeneration of cells and thus improve immune function and accelerate recovery.


Subject(s)
Biophysical Phenomena/physiology , COVID-19/metabolism , COVID-19/therapy , Energy Metabolism/physiology , Low-Level Light Therapy/methods , Ultrasonic Therapy/methods , COVID-19/immunology , Humans , Low-Level Light Therapy/trends , Ultrasonic Therapy/trends
SELECTION OF CITATIONS
SEARCH DETAIL