Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Acta Neurol Taiwan ; 32(2): 74-78, 2023 Jun 30.
Article in English | MEDLINE | ID: covidwho-2325741

ABSTRACT

BACKGROUND: During corona virus pandemic, various neurological complications of COVID-19 have been reported. Recent studies demonstrated different pathophysiology for neurological manifestations of COVID-19 such as mitochondrial dysfunction and damage to cerebral vasculature. In addition, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is a mitochondrial disorder with a variety of neurological symptoms. In this study, we aim to assess a potential predisposition in mitochondrial dysfunction of COVID-19, leading to MELAS presentation. METHODS: We studied three previously healthy patients with the first presentation of acute stroke-like symptoms, following COVID-19 infection. We analyzed the patients' clinical data and brain magnetic resonance imaging (MRI) lesions that presented to the neurological center of a university-affiliated hospital in Tehran, Iran, from September 2020 to August 2021. RESULTS: All cases are characterized by a temporoparietal abnormality in imaging studies and electroencephalogram (EEG). Based on electrodiagnostic tests, three patients were diagnosed with myopathy. In two brothers with relatively the same symptoms, one performed muscle biopsy finding myopathic process, and genetic testing confirmed a 3243A>G point mutation in a heteroplasmic state in one of our patients. CONCLUSION: Although MELAS is not a prevalent condition, the recent increase in the number of these patients in our center might indicate the potential role of COVID-19 in triggering the silent pre- existing mitochondrial dysfunction in these patients.


Subject(s)
Acidosis, Lactic , COVID-19 , MELAS Syndrome , Nervous System Diseases , Stroke , Male , Humans , MELAS Syndrome/complications , MELAS Syndrome/genetics , MELAS Syndrome/diagnosis , COVID-19/complications , COVID-19/pathology , Iran , Acidosis, Lactic/complications , Acidosis, Lactic/pathology , Stroke/etiology , Nervous System Diseases/complications , Nervous System Diseases/pathology , Mitochondria/pathology
2.
Mol Cell Biochem ; 2023 Apr 24.
Article in English | MEDLINE | ID: covidwho-2305431

ABSTRACT

Growth differentiation factor 15 (GDF15), a member of the transforming growth factor-beta superfamily, is expressed in several human organs. In particular, it is highly expressed in the placenta, prostate, and liver. The expression of GDF15 increases under cellular stress and pathological conditions. Although numerous transcription factors directly up-regulate the expression of GDF15, the receptors and downstream mediators of GDF15 signal transduction in most tissues have not yet been determined. Glial cell-derived neurotrophic factor family receptor α-like protein was recently identified as a specific receptor that plays a mediating role in anorexia. However, the specific receptors of GDF15 in other tissues and organs remain unclear. As a marker of cell stress, GDF15 appears to exert different effects under different pathological conditions. Cell senescence may be an important pathogenetic process and could be used to assess the progression of various lung diseases, including COVID-19. As a key member of the senescence-associated secretory phenotype protein repertoire, GDF15 seems to be associated with mitochondrial dysfunction, although the specific molecular mechanism linking GDF15 expression with ageing remains to be elucidated. Here, we focus on research progress linking GDF15 expression with the pathogenesis of various chronic lung diseases, including neonatal bronchopulmonary dysplasia, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and pulmonary hypertension, suggesting that GDF15 may be a key biomarker for diagnosis and prognosis. Thus, in this review, we aimed to provide new insights into the molecular biological mechanism and emerging clinical data associated with GDF15 in lung-related diseases, while highlighting promising research and clinical prospects.

3.
Pathogens ; 10(11)2021 Oct 21.
Article in English | MEDLINE | ID: covidwho-2279464

ABSTRACT

BACKGROUND: The causes and pathophysiological mechanisms of building-related symptoms (BRS) remain open. OBJECTIVE: We aimed to investigate the association between teachers' individual work-related symptoms and intrinsic in vitro toxicity in classrooms. This is a further analysis of a previously published dataset. METHODS: Teachers from 15 Finnish schools in Helsinki responded to the symptom survey. The boar sperm motility inhibition assay, a sensitive indicator of mitochondrial dysfunction, was used to measure the toxicity of wiped dust and cultured microbial fallout samples collected from the teachers' classrooms. RESULTS: 231 teachers whose classroom toxicity data had been collected responded to the questionnaire. Logistic regression analysis adjusted for age, gender, smoking, and atopy showed that classroom dust intrinsic toxicity was statistically significantly associated with the following 12 symptoms reported by teachers (adjusted ORs in parentheses): nose stuffiness (4.1), runny nose (6.9), hoarseness (6.4), globus sensation (9.0), throat mucus (7.6), throat itching (4.4), shortness of breath (12.2), dry cough (4.7), wet eyes (12.7), hypersensitivity to sound (7.9), difficulty falling asleep (7.6), and increased need for sleep (7.7). Toxicity of cultured microbes was found to be associated with nine symptoms (adjusted ORs in parentheses): headache (2.3), nose stuffiness (2.2), nose dryness (2.2), mouth dryness (2.8), hoarseness (2.2), sore throat (2.8), throat mucus (2.3), eye discharge (10.2), and increased need for sleep (3.5). CONCLUSIONS: The toxicity of classroom dust and airborne microbes in boar sperm motility inhibition assay significantly increased teachers' risk of work-related respiratory and ocular symptoms. Potential pathophysiological mechanisms of BRS are discussed.

4.
J Med Virol ; 95(2): e28553, 2023 02.
Article in English | MEDLINE | ID: covidwho-2267525

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) caused global pandemic and drastically affected the humankind. Mitochondrial mutations have been found to be associated with several respiratory diseases. Missense mutation and pathogenic mitochondrial variants might unveil the potential involvement of the mitochondrial genome in coronavirus disease 2019 (COVID-19) pathogenesis. The present study aims to elucidate the role of mitochondrial DNA (mtDNA) mutations, mitochondrial haplogroup, and energy metabolism in disease severity. The study was performed on 58 subjects comprising COVID-19-positive (n = 42) and negative (n = 16) individuals. COVID-19-positive subjects were further categorized into severe deceased (SD), severe recovered (SR), moderate (Mo), and mild (Mi) patients, while COVID-19-negative subjects were healthy control (HC) for the study. High throughput next-generation sequencing was done to investigate mtDNA mutations and haplogroups. The computational approach was applied to study the effect of mtDNA mutations on protein secondary structure. Real time polymerase chain reaction was used for mtDNA copy number determination and mitochondrial function parameters were also analyzed. We found 15 mtDNA mutations in MT-ND5, MT-ND4, MT-ND2, and MT-COI genes uniquely associated with COVID-19 severity affecting the secondary structure of proteins in COVID-19-positive subjects. Haplogroup analysis suggests that mtDNA haplogroups M3d1a and W3a1b might be potentially associated with COVID-19 pathophysiology. The mitochondrial function parameters were significantly altered in severe patients (SD and SR; p < 0.05). No significant relationship was found between mtDNA mutations and oxidative stress markers (p > 0.05). The study highlights the importance of mitochondrial reprogramming in COVID-19 patients and may provide a feasible approach toward finding a path for therapeutic interventions to COVID-19 disease.


Subject(s)
COVID-19 , Humans , COVID-19/pathology , SARS-CoV-2/genetics , Mutation , DNA, Mitochondrial/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology
5.
J Clin Endocrinol Metab ; 107(7): 1799-1811, 2022 06 16.
Article in English | MEDLINE | ID: covidwho-2288051

ABSTRACT

Obesity is associated with an increase in morbidity and mortality from coronavirus disease 2019 (COVID-19). The risk is related to the cytokine storm, a major contributor to multiorgan failure and a pathological character of COVID-19 patients with obesity. While the exact cause of the cytokine storm remains elusive, disorders in energy metabolism has provided insights into the mechanism. Emerging data suggest that adipose tissue in obesity contributes to the disorders in several ways. First, adipose tissue restricts the pulmonary function by generation of mechanical pressures to promote systemic hypoxia. Second, adipose tissue supplies a base for severe acute respiratory syndrome coronavirus 2 entry by overexpression of viral receptors [angiotensin-converting enzyme 2 and dipeptidyl peptidase 4]. Third, impaired antiviral responses of adipocytes and immune cells result in dysfunction of immunologic surveillance as well as the viral clearance systems. Fourth, chronic inflammation in obesity contributes to the cytokine storm by secreting more proinflammatory cytokines. Fifth, abnormal levels of adipokines increase the risk of a hyperimmune response to the virus in the lungs and other organs to enhance the cytokine storm. Mitochondrial dysfunction in adipocytes, immune cells, and other cell types (endothelial cells and platelets, etc) is a common cellular mechanism for the development of cytokine storm, which leads to the progression of mild COVID-19 to severe cases with multiorgan failure and high mortality. Correction of energy surplus through various approaches is recommended in the prevention and treatment of COVID-19 in the obese patients.


Subject(s)
Adipose Tissue , COVID-19 , Obesity , Adipose Tissue/metabolism , COVID-19/complications , Cytokine Release Syndrome , Cytokines/metabolism , Endothelial Cells/metabolism , Humans , Obesity/complications
6.
Nitric Oxide ; 133: 18-21, 2023 04 01.
Article in English | MEDLINE | ID: covidwho-2232939

ABSTRACT

Several COVID-19 patients frequently experience with happy hypoxia. Sometimes, the level of nitric oxide (NO) in COVID-19 patients was found to be greater than in non-COVID-19 hypoxemics and most of the cases lower. Induced or inhaled NO has a long history of usage as a therapy for hypoxemia. Excessive production of ROS and oxidative stress lower the NO level and stimulates mitochondrial malfunction is the primary cause of hypoxia-mediated mortality in COVID-19. Higher level of NO in mitochondria also the cause of dysfunction, because, excess NO can also diffuse quickly into mitochondria or through mitochondrial nitric oxide synthase (NOS). A precise dose of NO may increase oxygenation while also acting as an effective inhibitor of cytokine storm. NOS inhibitors may be used in conjunction with iNO therapy to compensate for the patient's optimal NO level. NO play a key role in COVID-19 happy hypoxia and a crucial component in the COVID-19 pathogenesis that demands a reliable and easily accessible biomarker to monitor.


Subject(s)
COVID-19 , Nitric Oxide , Humans , Nitric Oxide/pharmacology , COVID-19/complications , Hypoxia/drug therapy , Nitric Oxide Synthase , Mitochondria , Administration, Inhalation
7.
Mol Med ; 29(1): 13, 2023 01 26.
Article in English | MEDLINE | ID: covidwho-2214525

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS), a life-threatening condition during critical illness, is a common complication of COVID-19. It can originate from various disease etiologies, including severe infections, major injury, or inhalation of irritants. ARDS poses substantial clinical challenges due to a lack of etiology-specific therapies, multisystem involvement, and heterogeneous, poor patient outcomes. A molecular comparison of ARDS groups holds the potential to reveal common and distinct mechanisms underlying ARDS pathogenesis. METHODS: We performed a comparative analysis of urine-based metabolomics and proteomics profiles from COVID-19 ARDS patients (n = 42) and bacterial sepsis-induced ARDS patients (n = 17). To this end, we used two different approaches, first we compared the molecular omics profiles between ARDS groups, and second, we correlated clinical manifestations within each group with the omics profiles. RESULTS: The comparison of the two ARDS etiologies identified 150 metabolites and 70 proteins that were differentially abundant between the two groups. Based on these findings, we interrogated the interplay of cell adhesion/extracellular matrix molecules, inflammation, and mitochondrial dysfunction in ARDS pathogenesis through a multi-omic network approach. Moreover, we identified a proteomic signature associated with mortality in COVID-19 ARDS patients, which contained several proteins that had previously been implicated in clinical manifestations frequently linked with ARDS pathogenesis. CONCLUSION: In summary, our results provide evidence for significant molecular differences in ARDS patients from different etiologies and a potential synergy of extracellular matrix molecules, inflammation, and mitochondrial dysfunction in ARDS pathogenesis. The proteomic mortality signature should be further investigated in future studies to develop prediction models for COVID-19 patient outcomes.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Sepsis , Humans , COVID-19/complications , Proteomics , Multiomics , Respiratory Distress Syndrome/etiology , Sepsis/complications , Inflammation
8.
Eur J Pharmacol ; 940: 175475, 2023 Feb 05.
Article in English | MEDLINE | ID: covidwho-2165260

ABSTRACT

Vascular endothelial dysfunction plays a central role in the most dreadful human diseases, including stroke, tumor metastasis, and the coronavirus disease 2019 (COVID-19). Strong evidence suggests that angiotensin II (Ang II)-induced mitochondrial dysfunction is essential for endothelial dysfunction pathogenesis. However, the precise molecular mechanisms remain obscure. Here, polymerase-interacting protein 2 (Poldip 2) was found in the endothelial mitochondrial matrix and no effects on Poldip 2 and NADPH oxidase 4 (NOX 4) expression treated by Ang II. Interestingly, we first found that Ang II-induced NOX 4 binds with Poldip 2 was dependent on cyclophilin D (CypD). CypD knockdown (KD) significantly inhibited the binding of NOX 4 to Poldip 2, and mitochondrial ROS generation in human umbilical vein endothelial cells (HUVECs). Similar results were also found in cyclosporin A (CsA) treated HUVECs. Our previous study suggested a crosstalk between extracellular regulated protein kinase (ERK) phosphorylation and CypD expression, and gallic acid (GA) inhibited mitochondrial dysfunction in neurons depending on regulating the ERK-CypD axis. Here, we confirmed that GA inhibited Ang II-induced NOX 4 activation and mitochondrial dysfunction via ERK/CypD/NOX 4/Poldip 2 pathway, which provide novel mechanistic insight into CypD act as a key regulator of the NOX 4/Poldip 2 axis in Ang II-induced endothelial mitochondrial dysfunction and GA might be beneficial in the treatment of wide variety of diseases, such as COVID-19, which is worthy further research.


Subject(s)
COVID-19 , Vascular Diseases , Humans , NADPH Oxidase 4/metabolism , Angiotensin II/pharmacology , Angiotensin II/metabolism , Reactive Oxygen Species/metabolism , Cyclophilin D/metabolism , Cyclophilin D/pharmacology , NADPH Oxidases/metabolism , Oxidative Stress , Gallic Acid/pharmacology , COVID-19/metabolism , Mitochondria , Human Umbilical Vein Endothelial Cells
9.
Int J Mol Sci ; 23(23)2022 Nov 26.
Article in English | MEDLINE | ID: covidwho-2123706

ABSTRACT

The epithelial barrier's primary role is to protect against entry of foreign and pathogenic elements. Both COVID-19 and Inflammatory Bowel Disease (IBD) show commonalities in symptoms and treatment with sensitization of the epithelial barrier inviting an immune response. In this study we use a multi-omics strategy to identify a common signature of immune disease that may be able to predict for more severe patient outcomes. Global proteomic approaches were applied to transcriptome and proteome. Further semi- and relative- quantitative targeted mass spectrometry methods were developed to substantiate the proteomic and metabolomics changes in nasal swabs from healthy, COVID-19 (24 h and 3 weeks post infection); serums from Crohn's disease patients (scored for epithelial leak), terminal ileum tissue biopsies (patient matched inflamed and non-inflamed regions, and controls). We found that the tryptophan/kynurenine metabolism pathway is a 'hub' regulator of canonical and non-canonical transcription, macrophage release of cytokines and significant changes in the immune and metabolic status with increasing severity and disease course. Significantly modified pathways include stress response regulator EIF2 signaling (p = 1 × 10-3); energy metabolism, KYNU (p = 4 × 10-4), WARS (p = 1 × 10-7); inflammation, and IDO activity (p = 1 × 10-6). Heightened levels of PARP1, WARS and KYNU are predictive at the acute stage of infection for resilience, while in contrast, levels remained high and are predictive of persistent and more severe outcomes in COVID disease. Generation of a targeted marker profile showed these changes in immune disease underlay resolution of epithelial barrier function and have the potential to define disease trajectory and more severe patient outcomes.


Subject(s)
COVID-19 , Inflammatory Bowel Diseases , Humans , Tryptophan/metabolism , Proteomics , Inflammatory Bowel Diseases/metabolism , Inflammation/genetics , Inflammation/metabolism , Transcriptome
10.
Clin Transl Med ; 12(9): e1025, 2022 09.
Article in English | MEDLINE | ID: covidwho-2027333

ABSTRACT

BACKGROUND: Patients with COVID-19 display a broad spectrum of manifestations from asymptomatic to life-threatening disease with dysregulated immune responses. Mechanisms underlying the detrimental immune responses and disease severity remain elusive. METHODS: We investigated a total of 137 APs infected with SARS-CoV-2. Patients were divided into mild and severe patient groups based on their requirement of oxygen supplementation. All blood samples from APs were collected within three weeks after symptom onset. Freshly isolated PBMCs were investigated for B cell subsets, their homing potential, activation state, mitochondrial functionality and proliferative response. Plasma samples were tested for cytokine concentration, and titer of Nabs, RBD-, S1-, SSA/Ro- and dsDNA-specific IgG. RESULTS: While critically ill patients displayed predominantly extrafollicular B cell activation with elevated inflammation, mild patients counteracted the disease through the timely induction of mitochondrial dysfunction in B cells within the first week post symptom onset. Rapidly increased mitochondrial dysfunction, which was caused by infection-induced excessive intracellular calcium accumulation, suppressed excessive extrafollicular responses, leading to increased neutralizing potency index and decreased inflammatory cytokine production. Patients who received prior COVID-19 vaccines before infection displayed significantly decreased extrafollicular B cell responses and mild disease. CONCLUSION: Our results reveal an immune mechanism that controls SARS-CoV-2-induced detrimental B cell responses and COVID-19 severity, which may have implications for viral pathogenesis, therapeutic interventions and vaccine development.


Subject(s)
COVID-19 , Viral Vaccines , B-Lymphocytes , COVID-19 Vaccines , Cytokines , Humans , Mitochondria , SARS-CoV-2 , Severity of Illness Index , Viral Vaccines/pharmacology
11.
Advanced Functional Materials ; 2022.
Article in English | Web of Science | ID: covidwho-2003585

ABSTRACT

The widespread use of broad-spectrum antimicrobials has accelerated their entry into aquatic environment, which in turn can adversely affect aquatic organisms and humans, especially in the COVID-19 outbreak and the post-pandemic era. For early detection and intervention of adverse effects, this study develops a new carbon nanoprobe (CNP) that can reveal the adverse effects of trace amount triclosan (TCS), a commonly used broad-spectrum antimicrobial (BSA), through a direct visualization method. CNP has excellent fluorescent properties and strong positive charges, which can be applied as fluorescent indicator and trapped in mitochondria by electrostatic attraction. The highly sensitive responsiveness of CNP to mitochondrial membrane potential ensures the visualization method can be used for monitoring the adverse effects of TCS. The trace amount TCS monitoring is achieved according to the decrease of fluorescence signal in mitochondria and the change of mitochondrial morphological structure from lines to dots. Moreover, monitoring TCS level in aquatic organisms of zebrafish is further realized. Compared with the morphological toxicity test, this visualizing strategy reveals the adverse effects in organisms under low-dose TCS exposure more sensitively. This developed highly sensitive nanoprobe is cruical for direct BSA monitoring and thus prevents the harm of BSA to aquatic organisms and humans.

12.
Neurosci Behav Physiol ; 52(4): 511-514, 2022.
Article in English | MEDLINE | ID: covidwho-1955993

ABSTRACT

Objective. To study the efficacy of courses of i.v., Cytoflavin in combination with the standard rehabilitation program for postcovid syndrome for correction of postcovid asthenia. Materials and methods. Follow-up investigations were carried out in 45 patients with postcovid syndrome at the second stage of rehabilitation. Patients were divided into two groups of comparable sex and age. The volume of lung damage was also similar in both groups, at 25-80%. The 24 patients making up the comparison group received standard postcovid rehabilitation: pulsed magnetotherapy, inhalation therapy, aeroionotherapy, infrared laser therapy, courses of aerobic training, rational psychotherapy, and successive drug therapy. The 21 patients of the study group additionally received intravenous Cytoflavin daily for 10 days. The dynamics of increases in scores on the Rehabilitation Routing Scale, the Hamilton Depression Rating Scale (HDRS), the Asthenic Status Scale, and the 6-minute walk test at admission and discharge were also monitored. Results and conclusions. Addition of courses of intravenous Cytoflavin to the complex rehabilitation program for postcovid syndrome significantly improved the general functional state of the body, decreased levels of depression and asthenization, and increased physical exercise tolerance.

13.
Life (Basel) ; 12(5)2022 Apr 29.
Article in English | MEDLINE | ID: covidwho-1875685

ABSTRACT

Apart from ATP generation, mitochondria are involved in a wide range of functions, making them one of the most prominent organelles of the human cell. Mitochondrial dysfunction is involved in the pathophysiology of several diseases, such as cancer, neurodegenerative diseases, cardiovascular diseases, and metabolic disorders. This makes it a target for a variety of therapeutics for the diagnosis and treatment of these diseases. The use of nanoparticles to target mitochondria has significant importance in modern times because they provide promising ways to deliver drug payloads to the mitochondria by overcoming challenges, such as low solubility and poor bioavailability, and also resolve the issues of the poor biodistribution of drugs and pharmacokinetics with increased specificity. This review assesses nanoparticle-based drug-delivery systems, such as liposomes, DQAsome, MITO-Porters, micelles, polymeric and metal nanocarriers, as well as quantum dots, as mitochondria-targeted strategies and discusses them as a treatment for mitochondrial disorders.

14.
Am J Respir Cell Mol Biol ; 67(3): 275-283, 2022 09.
Article in English | MEDLINE | ID: covidwho-1765223

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that is currently causing a pandemic and has been termed coronavirus disease (COVID-19). The elderly or those with preexisting conditions like diabetes, hypertension, coronary heart disease, chronic obstructive pulmonary disease, cerebrovascular disease, or kidney dysfunction are more likely to develop severe cases when infected. Patients with COVID-19 admitted to the ICU have higher mortality than non-ICU patients. Critical illness has consistently posed a challenge not only in terms of mortality but also in regard to long-term outcomes of survivors. Patients who survive acute critical illness including, but not limited to, pulmonary and systemic insults associated with acute respiratory distress syndrome, pneumonia, systemic inflammation, and mechanical ventilation, will likely suffer from post-ICU syndrome, a phenomenon of cognitive, psychiatric, and/or physical disability after treatment in the ICU. Post-ICU morbidity and mortality continue to be a cause for concern when considering large-scale studies showing 12-month mortality risks of 11.8-21%. Previous studies have demonstrated that multiple mechanisms, including cytokine release, mitochondrial dysfunction, and even amyloids, may lead to end-organ dysfunction in patients. We hypothesize that COVID-19 infection will lead to post-ICU syndrome via potentially similar mechanisms as other chronic critical illnesses and cause long-term morbidity and mortality in patients. We consider a variety of mechanisms and questions that not only consider the short-term impact of the COVID-19 pandemic but its long-term effects that may not yet be imagined.


Subject(s)
COVID-19 , Pandemics , Aged , Critical Care , Critical Illness , Humans , SARS-CoV-2
15.
National Technical Information Service; 2020.
Non-conventional in English | National Technical Information Service | ID: grc-753573

ABSTRACT

Prostate cancer (PCa) is the most common cancer among men. Androgen deprivation therapy (ADT)is the standard treatment for advanced and metastatic PCa and nearly 400,000 men remain on androgen deprivation therapy (ADT) for advanced PCa in the U.S. Unfortunately, ADT also induces a decrease in muscle mass and function, known as sarcopenia, a condition that leads to decreased endurance, increased fatigue, falls, poor health-related quality of life (HR-QOL) and increased mortality. The mechanisms underlying the development of ADT-induced sarcopenia are incompletely understood and remain a significant barrier to the development of therapies for this condition. Mitochondria play an essential role in generating the adenosine triphosphate (ATP) needed for muscle contraction and abnormalities in mitochondria function have been reported in animal models of sarcopenia. The extent to which mitochondrial dysfunction mediates ADT-induced sarcopenia and muscle dysfunction is not known. The overall goal of this proposal is to establish the role of mitochondrial dysfunction on ADT-induced sarcopenia in patients with PCa. Our hypothesis is that ADT in men with PCa will induce mitochondrial dysfunction leading to sarcopenia. To test this hypothesis, we will carry out a pilot study of men with PCa undergoing ADT (n=60).As of August 26, 2020, we have enrolled seventeen research participants in the study. Research participant recruitment and performance of study visits were impacted beginning in March due to the COVID-19 epidemic. We are following local VA and UW communications closely to ensure we meet their recommended guidelines for resuming human subjects research

16.
Medicina (Kaunas) ; 58(3)2022 Mar 12.
Article in English | MEDLINE | ID: covidwho-1742544

ABSTRACT

Dyspnea, shortness of breath, and chest pain are frequent symptoms of post-COVID syndrome (PCS). These symptoms are unrelated to organ damage in most patients after mild acute COVID infection. Hyperventilation has been identified as a cause of exercise-induced dyspnea in PCS. Since there is a broad overlap in symptomatology with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), causes for dyspnea and potential consequences can be deduced by a stringent application of assumptions made for ME/CFS in our recent review papers. One of the first stimuli of respiration in exercise is caused by metabolic feedback via skeletal muscle afferents. Hyperventilation in PCS, which occurs early on during exercise, can arise from a combined disturbance of a poor skeletal muscle energetic situation and autonomic dysfunction (overshooting respiratory response), both found in ME/CFS. The exaggerated respiratory response aggravating dyspnea does not only limit the ability to exercise but further impairs the muscular energetic situation: one of the buffering mechanisms to respiratory alkalosis is a proton shift from intracellular to extracellular space via the sodium-proton-exchanger subtype 1 (NHE1), thereby loading cells with sodium. This adds to two other sodium loading mechanisms already operative, namely glycolytic metabolism (intracellular acidosis) and impaired Na+/K+ATPase activity. High intracellular sodium has unfavorable effects on mitochondrial calcium and metabolism via sodium-calcium-exchangers (NCX). Mitochondrial calcium overload by high intracellular sodium reversing the transport mode of NCX to import calcium is a key driver for fatigue and chronification. Prevention of hyperventilation has a therapeutic potential by keeping intracellular sodium below the threshold where calcium overload occurs.


Subject(s)
COVID-19 , Fatigue Syndrome, Chronic , COVID-19/complications , Dyspnea/etiology , Exercise , Fatigue Syndrome, Chronic/etiology , Fatigue Syndrome, Chronic/therapy , Humans , Sodium
17.
Front Immunol ; 12: 799896, 2021.
Article in English | MEDLINE | ID: covidwho-1662583

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection results in rapid T lymphocytopenia and functional impairment of T cells. The underlying mechanism, however, remains incompletely understood. In this study, we focused on characterizing the phenotype and kinetics of T-cell subsets with mitochondrial dysfunction (MD) by multicolor flow cytometry and investigating the association between MD and T-cell functionality. While 73.9% of study subjects displayed clinical lymphocytopenia upon hospital admission, a significant reduction of CD4 or CD8 T-cell frequency was found in all asymptomatic, symptomatic, and convalescent cases. CD4 and CD8 T cells with increased MD were found in both asymptomatic and symptomatic patients within the first week of symptom onset. Lower proportion of memory CD8 T cell with MD was found in severe patients than in mild ones at the stage of disease progression. Critically, the frequency of T cells with MD in symptomatic patients was preferentially associated with CD4 T-cell loss and CD8 T-cell hyperactivation, respectively. Patients bearing effector memory CD4 and CD8 T cells with the phenotype of high MD exhibited poorer T-cell responses upon either phorbol 12-myristate-13-acetate (PMA)/ionomycin or SARS-CoV-2 peptide stimulation than those with low MD. Our findings demonstrated an MD-associated mechanism underlying SARS-CoV-2-induced T lymphocytopenia and functional impairment during the acute phase of infection.


Subject(s)
COVID-19/complications , Lymphopenia/complications , Lymphopenia/etiology , Mitochondrial Diseases/etiology , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , Female , Humans , Immunologic Memory/immunology , Ionomycin/therapeutic use , Lymphopenia/immunology , Male , Middle Aged , Mitochondria/immunology , Mitochondrial Diseases/immunology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/therapeutic use , Polymethacrylic Acids/therapeutic use , COVID-19 Drug Treatment
18.
Mediterranean Journal of Nutrition and Metabolism ; 14(4):401-416, 2021.
Article in English | Web of Science | ID: covidwho-1559639

ABSTRACT

BACKGROUND: Nowadays, medicinal plants have attracted great interest in treatment of human diseases. Rosemary is a well-known medicinal plant which has been widely used for different therapeutic purposes. METHODS: This is a narrative reviewusing databases including PubMed, ISI, Scopus, ScienceDirect, Cochrane, and google scholar, the most authoritative articles were searched, screened, and analyzed. RESULTS: Rosemary is a natural antioxidant which removes reactive oxygen species from tissues and increases expression on Nrf2 gene. Rosemary and its metabolites reduce inflammation by inhibiting production of pro-inflammatory cytokines, decreasing expression of NF-kappa B, inhibiting infiltration of immune cells to inflamed sites, and affecting gut microbiome. Besides, rosmarinic acid in rosemary extract has positive effects on renin-angiotensin-system. Rosemary affects respiratory system by reducing oxidative stress, inflammation, muscle spasm, and also through anti-fibrotic properties. Carnosic acid is able to penetrate blood-brain-barrier and act against free radicals, ischemia and neurodegeneration in brain. Cardioprotective effects include correcting lipid profile, controlling blood pressure by inhibition of ACE, prevention of atherosclerosis, and reduction of cardiac muscle hypertrophy. CONCLUSIONS: Accordingly, rosemary supplementation has potential protective effects against COVID-19 and other cytokine storm associated infections, a conclusion that needs more evaluations in the next clinical trials.

19.
Free Radic Res ; 55(9-10): 982-995, 2021 Oct.
Article in English | MEDLINE | ID: covidwho-1555313

ABSTRACT

The inflammation and activation of the immune system induced by SARS-CoV-2 are mediated by a pro-oxidant microenvironment that can induce cytotoxic effects that enhance tissue damage, favoring organic deterioration. We investigated whether the induction of oxidative stress and inflammation by COVID-19 infection could inhibit mitochondrial function and cause cellular damage in leukocytes. We evaluated levels of oxidative/inflammation markers and their correlation with mitochondrial function and leukocyte cell death in COVID-19 patients at two moments: viremia and severe sepsis with multi-organ failure. COVID-19 induces increased oxidative stress and inflammation markers that activate cellular damage processes. In the viremia stage, an increase in peroxide, nitric oxide, carbonylated proteins, and IL-6 was observed, which was correlated with a marked inhibition of mitochondrial function, decreased cell viability, early apoptosis, necrosis, and leukocytes-reactivity. The severe sepsis stage with multi-organ failure also showed a further increase in levels of peroxide, carbonylated proteins, and IL-6, with a slight decrease in nitric oxide. This oxidative process and inflammation were correlated with less inhibition of mitochondrial function, decreased cell viability and an increase in late apoptosis, and morphology changes evidencing damage in the leukocytes. SARS-CoV-2 induced damage promotes levels of oxidative stress and inflammation markers and mitochondrial dysfunction that potentiate morphological changes and cell death in leukocytes. These processes explain the rapid changes in the immune system, and that present an initial over-activation and early massive death due to SARS-CoV-2 infection, promoting endothelial-alveolar damage that would cause multi-organ failure, sustained by oxidative stress and inflammation.


Subject(s)
COVID-19 , Cell Death , Humans , Inflammation , Leukocytes , Mitochondria , Oxidative Stress , SARS-CoV-2
20.
J Neuroimmune Pharmacol ; 16(4): 770-784, 2021 12.
Article in English | MEDLINE | ID: covidwho-1446201

ABSTRACT

Emerging clinical data from the current COVID-19 pandemic suggests that ~ 40% of COVID-19 patients develop neurological symptoms attributed to viral encephalitis while in COVID long haulers chronic neuro-inflammation and neuronal damage result in a syndrome described as Neuro-COVID. We hypothesize that SAR-COV2 induces mitochondrial dysfunction and activation of the mitochondrial-dependent intrinsic apoptotic pathway, resulting in microglial and neuronal apoptosis. The goal of our study was to determine the effect of SARS-COV2 on mitochondrial biogenesis and to monitor cell apoptosis in human microglia non-invasively in real time using Raman spectroscopy, providing a unique spatio-temporal information on mitochondrial function in live cells. We treated human microglia with SARS-COV2 spike protein and examined the levels of cytokines and reactive oxygen species (ROS) production, determined the effect of SARS-COV2 on mitochondrial biogenesis and examined the changes in molecular composition of phospholipids. Our results show that SARS- COV2 spike protein increases the levels of pro-inflammatory cytokines and ROS production, increases apoptosis and increases the oxygen consumption rate (OCR) in microglial cells. Increases in OCR are indicative of increased ROS production and oxidative stress suggesting that SARS-COV2 induced cell death. Raman spectroscopy yielded significant differences in phospholipids such as Phosphatidylinositol (PI), phosphatidylserine (PS), phosphatidylethanolamine (PE) and phosphatidylcholine (PC), which account for ~ 80% of mitochondrial membrane lipids between SARS-COV2 treated and untreated microglial cells. These data provide important mechanistic insights into SARS-COV2 induced mitochondrial dysfunction which underlies neuropathology associated with Neuro-COVID.


Subject(s)
COVID-19 , Microglia , Humans , Mitochondrial Dynamics , Pandemics , RNA, Viral , SARS-CoV-2 , Spike Glycoprotein, Coronavirus
SELECTION OF CITATIONS
SEARCH DETAIL