1
|
Qaisar R, Karim A, Iqbal MS, Muhammad T, Ahmad F, Alkahtani SA. The Pathology of Intestinal Mucosal Disruption; Implications for Muscle Loss and Physical Dependency from Late Adolescence to Octogenarians. Int J Gen Med 2024; 17:6117-6126. [PMID: 39687221 PMCID: PMC11648551 DOI: 10.2147/ijgm.s501358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/07/2024] [Indexed: 12/18/2024] Open
Abstract
Background and Objectives A pathological increase in intestinal permeability causes muscle loss and physical decline by inducing systemic inflammation and oxidative stress. However, most relevant studies investigate older adults, and the appropriate data across age spans remain elusive. This study aimed to examine the associations of intestinal permeability with muscle loss and physical decline across a large span of ages. We measured plasma zonulin, a marker of increased intestinal permeability, from adolescents to octogenarians in association with muscle health and gait speed. Research Methods and Procedures In this cross-sectional, observational study, we recruited healthy men, including young (age=18-35 years, n=135), middle-aged (age=35-59 years, n=118), and older (age=60-90 years, n=163) adults for evaluating - handgrip strength (HGS), skeletal muscle mass index (SMI), and gait speed. We also measured plasma zonulin, c-reactive proteins (CRP), and 8-isoprostanes using ELISA assays. Results Plasma zonulin gradually increased from young and middle-aged to older adults (all p<0.05). Conversely, HGS and gait speed were progressively reduced from young and middle-aged to older adults (all p<0.05). In addition, older adults also exhibited lower SMI than young and middle-aged men (both p<0.05). Plasma zonulin exhibited significant negative correlations with HGS and gait speed and positive correlations with CRP and 8-isoprostanes in middle-aged and older men (all p<0.05). We also found significant areas under the curve for the efficacy of plasma zonulin in diagnosing low HGS (<27kg) and gait speed (0.8 m/s). After adjustment for age, plasma zonulin demonstrated robust negative correlations with HGS and gait speed and positive correlations with CRP and 8-isoprostanes in the cumulative cohort. Conclusion Altogether, an increasing intestinal leak from middle age onward contributes to muscle weakness and physical decline. Our data is clinically relevant in understanding and treating physical dependency in middle-aged and older adults.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Space Medicine Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Cardiovascular Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - M Shahid Iqbal
- Department of Neurology and Stroke Medicine, Rehman Medical Institute, Peshawar, 25124, Pakistan
| | - Tahir Muhammad
- Department of Biochemistry, Gomal Medical College, Dera Ismail Khan, Pakistan
| | - Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Space Medicine Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Cardiovascular Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Shaea A Alkahtani
- Exercise Physiology Department, College of Sport Sciences and Physical Activity, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
2
|
Qaisar R, Javed M, Khan IM, Ahmad F, Karim A. Metformin improves skeletal muscle and physical capacity by stabilizing neuromuscular junction in older adults. Arch Gerontol Geriatr 2024; 127:105587. [PMID: 39084174 DOI: 10.1016/j.archger.2024.105587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVES Metformin is an anti-diabetic drug with protective effects on skeletal muscle and physical capacity. However, the relevant mechanisms of action on skeletal muscle remain poorly understood. We investigated the potential contribution of neuromuscular junction (NMJ) degradation to skeletal muscle and physical capacity in geriatric men taking metformin. METHOD We recruited geriatric men for placebo (Age=73.1 ± 4.2 years, n = 70) and metformin (Age=70.1 ± 4.5 years, n = 62) groups. The patients in the metformin group received 1700 mg of metformin twice a day for 16 weeks. We measured plasma c-terminal agrin-fragment-22 (CAF22) and neurofilament light chain (NfL) as markers of neuromuscular junction (NMJ) degradation and neurodegeneration, respectively, with relevance to handgrip strength (HGS) and short physical performance battery (SPPB; a marker of physical capacity) in older adults taking metformin. These findings were associated with reduced oxidative stress in the metformin group. RESULTS At baseline, both groups had similar HGS, gait speed, SPPB scores, and plasma biochemistry. Metformin improved HGS, gait speed, and cumulative SPPB scores in geriatric men (all p < 0.05). Metformin also reduced plasma CAF22 and NfL levels when compared to baseline. Similar observations were not found in the placebo group. Correlation analysis revealed significant correlations of plasma CAF22 with HGS, gait speed, and cumulative SPPB scores in the metformin group. These observations were associated with reduced oxidative stress in the metformin group. CONCLUSION Altogether, the restorative effects of metformin on skeletal muscle and physical capacity involve NMJ stabilization. Our data is clinically relevant for geriatric men with functional disabilities.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Mashal Javed
- Department of Physiotherapy, Burn and Plastic Surgery Center, Hayatabad Medical Complex, Hayatabad, Peshawar 25000, Khyber Pakhtunkhwa, Pakistan
| | - Imran Muhammad Khan
- Burn and Plastic Surgery Center, Hayatabad Medical Complex, Hayatabad, Peshawar 25000, Khyber Pakhtunkhwa, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
3
|
Siemińska I, Bukowska-Strakova K, Surmiak M, Ptak K, Szymońska I, Olchawa-Czech A, Mól N, Błyszczuk P, Sanak M, Baran J, Kwinta P, Siedlar M. Cytokine landscape in hospitalized children with multisystem inflammatory syndrome. Sci Rep 2024; 14:22803. [PMID: 39354098 PMCID: PMC11445419 DOI: 10.1038/s41598-024-73956-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
The etiology of multisystem inflammatory syndrome in children (MIS-C), frequently observed following COVID-19 infection, remains elusive. This study unveils insights derived from cytokine analysis in the sera of MIS-C patients, both before and after the administration of intravenous immunoglobulin (IVIG) and glucocorticosteroids (GCS). In this study, we employed a comprehensive 45-cytokine profile encompassing a spectrum of widely recognized proinflammatory and antiinflammatory cytokines, as well as growth factors, along with other soluble mediators. The analysis delineates three principal cytokine-concentration patterns evident in the patients' sera. Pattern no.1 predominantly features proinflammatory cytokines (IL-6, IL-15, IL-1ra, granulocyte-macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor α (TNFα), C-X-C motif chemokine ligand 10 (CXCL10/ IP-10), and IL-10) exhibiting elevated concentrations upon admission, swiftly normalizing post-hospital treatment. Pattern no. 2 includes cytokines (IL-17 A, IL-33, IFNγ, vascular endothelial growth factor (VEGF), and programmed death ligand (PD-L1)) with moderately elevated levels at admission, persisting over 7-10 days of hospitalization despite the treatment. Pattern no. 3 comprises cytokines which concentrations escalated after 7-10 days of hospitalization and therapy, including IL-1α, IL-1β, IL-2, IL-13, platelet-derived growth factor AA/BB (PDGF AA/BB). The observed in cytokine profile of MIS-C patients showed a transition from acute inflammation to sustaining inflammation which turned into induction of humoral memory mechanisms and various defense mechanisms, contributing to recovery.
Collapse
Affiliation(s)
- Izabela Siemińska
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
- Institute of Veterinary Sciences, University Center of Veterinary Medicine JU-AU, University of Agriculture in Kraków, al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - Karolina Bukowska-Strakova
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
| | - Marcin Surmiak
- Department of Internal Medicine, Jagiellonian University Medical College, Skawinska 8, Krakow, 31-066, Poland
| | - Katarzyna Ptak
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
| | - Izabela Szymońska
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
| | - Anna Olchawa-Czech
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
| | - Nina Mól
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
| | - Przemysław Błyszczuk
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Skawinska 8, Krakow, 31-066, Poland
| | - Jarek Baran
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland
| | - Przemko Kwinta
- Department of Paediatrics, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland.
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, Krakow, 30-663, Poland.
| |
Collapse
|
4
|
Gavrilova NY, Normatov MG, Soprun LA, Utekhin VJ, Fedotkina TV, Churilov LP. Autoantigens of Small Nerve Fibers and Human Coronavirus Antigens: Is There a Possibility for Molecular Mimicry? Curr Microbiol 2024; 81:366. [PMID: 39297982 DOI: 10.1007/s00284-024-03885-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/05/2024] [Indexed: 09/21/2024]
Abstract
In post-COVID-19 syndrome, clinical presentation of the nerve fiber dysfunction plays an important role. The possibility of autoantigen cross-mimicry of human coronaviruses and the peripheral nervous system needs to be investigated. The bioinformatic analysis was applied to search for possible common protein sequences located in the immunoreactive epitopes. Among the autoantigens of the human nervous system, fibroblast growth factor receptor protein 3, myelin protein P0, myelin protein P2, sodium channel protein type 9, alpha protein subunit, plexin-D1 protein and ubiquitin-carboxyl-terminal hydrolase protein of the L1 isoenzyme were selected. The original "Alignmentaj" analytical program was created. The UniProt database, Protein Data Bank, and AlphaFold databases were used. The analysis of protein sequence similarities of spike glycoproteins in human coronaviruses revealed common pentapeptides of the MERS-CoV-2 virus with the fibroblast growth factor receptor 3 and myelin protein P2. Among seasonal coronaviruses, common peptide sequences were identified in HCoV-HKU-1 virus with sodium channel protein type 9 subunit alpha and Plexin-D1, HCoV-OC43 with Plexin-D1, as well as HCoV-NL63 with Plexin-D1 and Ubiquitin carboxyl-terminal hydrolase isozyme L1. Some shared peptides belong to immunoreactive epitopes. The most important targets for the molecular similarities are the sodium channel subunits and fibroblast growth factor receptor 3, both for seasonal and highly pathogenic coronaviruses. The data obtained make it possible to identify new potential targets for the development of autoimmune reactions that may occur against the background of the infections with highly pathogenic as well as seasonal coronaviruses.
Collapse
Affiliation(s)
- Natalia Y Gavrilova
- The Laboratory of the Mosaic of Autoimmunity and Department of Pathology, Saint Petersburg State University, 199034, Saint-Petersburg, Russia
- Saint Petersburg State University Hospital, 198103, Saint-Petersburg, Russia
| | - Muslimbek G Normatov
- The Laboratory of the Mosaic of Autoimmunity and Department of Pathology, Saint Petersburg State University, 199034, Saint-Petersburg, Russia.
| | - Lidiya A Soprun
- The Laboratory of the Mosaic of Autoimmunity and Department of Pathology, Saint Petersburg State University, 199034, Saint-Petersburg, Russia
- Saint Petersburg State University Hospital, 198103, Saint-Petersburg, Russia
| | - Vladimir J Utekhin
- The Laboratory of the Mosaic of Autoimmunity and Department of Pathology, Saint Petersburg State University, 199034, Saint-Petersburg, Russia
- The Department of Pathophysiology with the Course of Immunopathology, Saint Petersburg State Pediatric Medical University, 194100, Saint-Petersburg, Russia
| | - Tamara V Fedotkina
- Almazov National Medical Research Centre, 197341, Saint-Petersburg, Russia
| | - Leonid P Churilov
- The Laboratory of the Mosaic of Autoimmunity and Department of Pathology, Saint Petersburg State University, 199034, Saint-Petersburg, Russia
| |
Collapse
|
5
|
Qaisar R, Karim A, Iqbal MS, Ahmad F, Hussain MA. Tracking the Plasma C-Terminal Agrin Fragment as a Biomarker of Neuromuscular Decline in 18- to 87-Year-Old Men. Mol Diagn Ther 2024; 28:611-620. [PMID: 38961032 DOI: 10.1007/s40291-024-00724-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVES Plasma C-terminal agrin-fragment-22 (CAF22), a breakdown product of neuromuscular junction, is a potential biomarker of muscle loss. However, its levels from adolescence to octogenarians are unknown. METHODS We evaluated young (18-34 years, n = 203), middle-aged (35-59 years, n = 163), and old men (60-87 years, n = 143) for CAF22, handgrip strength (HGS), appendicular skeletal-mass index (ASMI), and gait speed. RESULTS We found an age-associated increase in CAF22 from young (100.9 ± 29 pmol) to middle-aged (128.3 ± 38.7 pmol) and older men (171.5 ± 35.5 pmol) (all p<0.05). This was accompanied by a gradual reduction in HGS (37.7 ± 6.1 kg, 30.2 ± 5.2 kg, and 26.6 ± 4.7 kg, for young, middle-aged, and old men, respectively), ASMI (8.02 ± 1.02 kg/m2, 7.65 ± 0.92 kg/m2, 6.87 ± 0.93 kg/m2, for young, middle-aged, and old men, respectively), and gait speed (1.29 ± 0.24 m/s, 1.05 ± 0.16 m/s, and 0.81 ± 0.13 m/s, for young, middle-aged, and old men, respectively). After adjustment for age, we found negative regressions of CAF22 with HGS (- 0.0574, p < 0.001) and gait speed (- 0.0162, p < 0.001) in the cumulative cohort. The receiver operating characteristics analysis revealed significant efficacy of plasma CAF22 in diagnosing muscle weakness (HGS < 27 kg) (middle-aged men; AUC = 0.731, 95% CI = 0.629-0.831, p < 0.001, Older men; AUC = 0.816, 95% CI = 0.761-0.833, p < 0.001), and low gait speed (0.8 m/s) (middle-aged men; AUC = 0.737, 95% CI = 0.602-0.871, p < 0.001, older men; AUC = 0.829, 95% CI = 0.772-0.886, p < 0.001), and a modest efficacy in diagnosing sarcopenia (middle-aged men; AUC = 0.701, 95% CI = 0.536-0.865, p = 0.032, older men; AUC = 0.822, 95% CI = 0.759-0.884, p < 0.001) in middle-aged and older men. CONCLUSION Altogether, CAF22 increases with advancing age and may be a reliable marker of muscle weakness and low gait speed.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Space Medicine Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates.
- Cardiovascular Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates.
| | - Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - M Shahid Iqbal
- Department of Neurology and Stroke Medicine, Rehman Medical Institute, Peshawar, 25124, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Space Medicine Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - M Azhar Hussain
- Department of Finance and Economics, College of Business Administration, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Department of Social Sciences and Business, Roskilde University, 4000, Roskilde, Denmark
| |
Collapse
|
6
|
Kenny G, Saini G, Gaillard CM, Negi R, Alalwan D, Garcia Leon A, McCann K, Tinago W, Kelly C, Cotter AG, de Barra E, Horgan M, Yousif O, Gautier V, Landay A, McAuley D, Feeney ER, O'Kane C, Mallon PWG. Early inflammatory profiles predict maximal disease severity in COVID-19: An unsupervised cluster analysis. Heliyon 2024; 10:e34694. [PMID: 39144942 PMCID: PMC11320140 DOI: 10.1016/j.heliyon.2024.e34694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Background The inflammatory changes that underlie the heterogeneous presentations of COVID-19 remain incompletely understood. In this study we aimed to identify inflammatory profiles that precede the development of severe COVID-19, that could serve as targets for optimised delivery of immunomodulatory therapies and provide insights for the development of new therapies. Methods We included individuals sampled <10 days from COVID-19 symptom onset, recruited from both inpatient and outpatient settings. We measured 61 biomarkers in plasma, including markers of innate immune and T cell activation, coagulation, tissue repair and lung injury. We used principal component analysis and hierarchical clustering to derive biomarker clusters, and ordinal logistic regression to explore associations between cluster membership and maximal disease severity, adjusting for known risk factors for severe COVID-19. Results In 312 individuals, median (IQR) 7 (4-9) days from symptom onset, we found four clusters. Cluster 1 was characterised by low overall inflammation, cluster 2 was characterised by higher levels of growth factors and markers of endothelial activation (EGF, VEGF, PDGF, TGFα, PAI-1 and p-selectin). Cluster 3 and 4 both had higher overall inflammation. Cluster 4 had the highest levels of most markers including markers of innate immune activation (IL6, procalcitonin, CRP, TNFα), and coagulation (D-dimer, TPO), in contrast cluster 3 had the highest levels of alveolar epithelial injury markers (RAGE, ST2), but relative downregulation of growth factors and endothelial activation markers, suggesting a dysfunctional inflammatory pattern. In unadjusted and adjusted analysis, compared to cluster 1, cluster 3 had the highest odds of progressing to more severe disease (unadjusted OR (95%CI) 9.02 (4.53-17.96), adjusted OR (95%CI) 6.02 (2.70-13.39)). Conclusion Early inflammatory profiles predicted subsequent maximal disease severity independent of risk factors for severe COVID-19. A cluster with downregulation of growth factors and endothelial activation markers, and early evidence of alveolar epithelial injury, had the highest risk of severe COVID-19.
Collapse
Affiliation(s)
- Grace Kenny
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Gurvin Saini
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Colette Marie Gaillard
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Riya Negi
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Dana Alalwan
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Alejandro Garcia Leon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Kathleen McCann
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Willard Tinago
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Christine Kelly
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Aoife G. Cotter
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Eoghan de Barra
- Department of International Health and Tropical Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mary Horgan
- Department of Infectious Diseases, Cork University Hospital, Wilton, Cork, Ireland
| | - Obada Yousif
- Department of Endocrinology, Wexford General Hospital, Wexford, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Alan Landay
- Department of Internal Medicine, Rush University, Chicago, IL, USA
| | | | - Eoin R. Feeney
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| | | | - Patrick WG. Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
7
|
Qaisar R, Burki A, Karim A, Ustrana S, Ahmad F. The Association of Intestinal Leak with Sarcopenia and Physical Disability in Patients with Various Stages of Chronic Kidney Disease. Calcif Tissue Int 2024; 115:132-141. [PMID: 38829421 DOI: 10.1007/s00223-024-01233-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024]
Abstract
Sarcopenia is related to disease severity in chronic kidney disease (CKD) patients; however, its pathophysiology remains poorly known. We investigated the associations of biomarkers of intestinal leak with sarcopenia in various stages of CKD. We recruited 61-76-year-old male controls and patients with various stages of CKD (n = 36-57/group) for measuring plasma lipopolysaccharide-binding protein (LBP) and zonulin (markers of intestinal leak), handgrip strength (HGS), skeletal mass index (SMI), and gait speed (markers of sarcopenia), and short physical performance battery (SPPB; marker of physical capacity). CKD stages 4 and 5 were associated with lower HGS, SMI, gait speed, and cumulative SPPB scores and a higher sarcopenia prevalence than controls and patients with CKD stages 1 and 2 (all p < 0.05). CKD patients (stages 1 and 2) had elevated plasma zonulin and LBP when compared with CKD stages 4 and 5. Plasma zonulin and LBP exhibited significant correlations with renal function, HGS, gait speed, SPPB scores, and oxidative stress markers in CKD stages 4 and 5 (all p < 0.05). However, similar relations were not found in early CKD. Collectively, intestinal leak may be contributing to sarcopenia and physical disability in the advanced stages of CKD.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| | - Ayousha Burki
- Department of Nephrology, Divisional Headquarter Hospital, Gomal Medical College, Dera Ismail Khan, Pakistan
| | - Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Iron Biology Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Shahjahan Ustrana
- Department of Biochemistry, Gomal Medical College, Dera Ismail Khan, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
8
|
Qaisar R, Karim A, Muhammad T, Iqbal MS, Ahmad F. Metformin Improves Sarcopenia-Related Quality of Life in Geriatric Adults: A Randomized Controlled Trial. Arch Med Res 2024; 55:102998. [PMID: 38615625 DOI: 10.1016/j.arcmed.2024.102998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/23/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
OBJECTIVES Metformin protects against age-related muscle decline, termed sarcopenia. However, the effects on sarcopenia quality-of-life (SarQoL) are unknown. We investigated the effects of metformin on SarQoL and associated mechanisms in older adults. METHOD This double-blind randomized, placebo-controlled trial included geriatric adult men, divided into non-sarcopenic controls (age = 72.2 ± 4.3 years, n = 52) and two groups of patients with sarcopenia randomized into placebo (age at baseline = 74.4 ± 5.7 years, n = 54) and metformin (age at baseline = 71.2 ± 3.9 years, n = 47) groups. Patients in the metformin group received 1.7 grams twice daily for four months. We evaluated SarQoL, handgrip strength (HGS), plasma zonulin, c-reactive protein (CRP), and 8-isoprostanes. RESULTS Patients with sarcopenia had lower HGS and SarQoL than controls (both p <0.05). Metformin improved the HGS and the SarQoL domains related to physical and mental health, locomotion, and leisure activities, as well as cumulative SarQoL scores (all p <0.05). Metformin also prevented the decline in the SarQoL domains for functionality and fear. Among plasma biomarkers, metformin reduced the levels of zonulin, CRP, 8-isoprostanes, and creatine kinase. We also found a significant correlation of plasma zonulin with cumulative SarQoL in patients with sarcopenia taking metformin, suggesting a role for intestinal repair in improving SarQoL. Finally, metformin did not affect body composition and gait speed. CONCLUSION Overall, metformin improved HGS and SarQoL by repairing intestinal leakage. Our data have clinical relevance for improving the quality of life in older adults with sarcopenia.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Iron Biology Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Tahir Muhammad
- Department of Biochemistry, Gomal Medical College, Dera Ismail Khan, Pakistan
| | - M Shahid Iqbal
- Department of Neurology and Stroke Medicine, Rehman Medical Institute, Peshawar, Pakistan
| | - Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
9
|
Qaisar R, Burki A, Karim A, Iqbal MS, Ahmad F. Probiotics Supplements Improve the Sarcopenia-Related Quality of Life in Older Adults with Age-Related Muscle Decline. Calcif Tissue Int 2024; 114:583-591. [PMID: 38642090 DOI: 10.1007/s00223-024-01211-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/14/2024] [Indexed: 04/22/2024]
Abstract
A pathological increase in intestinal leak is implicated in age-associated muscle loss, termed sarcopenia, and reduced sarcopenia-related quality-of-life (SarQoL). However, the potential therapies remain elusive. We investigated the effects of probiotic supplementation on sarcopenia and SarQoL in geriatric older adults. We randomized sarcopenic men into placebo (age = 71.4 ± 3.9 years, n = 63) and probiotic (age = 73 ± 4.1 years, n = 60) groups for 16 weeks. The probiotic used was one capsule daily of Vivomix 112 billion for 16 weeks. We measured sarcopenia parameters of handgrip strength (HGS) and skeletal mass index (SMI), plasma zonulin (marker of the intestinal leak), and SarQoL using a targeted questionnaire. Probiotics improved the SarQoL scores for locomotion, functionality, and activities of daily living and prevented a decline in cumulative SarQoL observed in the placebo group (all p < 0.05). Probiotic supplementation also reduced plasma zonulin and marker of systemic bacterial load. These changes were accompanied by an increase in HGS and maintenance of gait speed in the probiotic group compared to the placebo group. Correlation analysis revealed significant associations of cumulative SarQoL scores with plasma zonulin and HGS in the probiotic group. Collectively, probiotics improved SarQoL and HGS by repairing pathological intestinal leak. Future studies may further dissect the relation between intestinal leak and SarQoL in older adults taking probiotics.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates.
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates.
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates.
| | - Ayousha Burki
- Department of Nephrology, Divisional Headquarter Teaching Hospital, Gomal Medical College, Dera Ismail Khan, 30130, Pakistan
| | - Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Iron Biology Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - M Shahid Iqbal
- Department of Neurology and Stroke Medicine, Rehman Medical Institute, Peshawar, 25120, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates
| |
Collapse
|
10
|
Qaisar R, Khan IM, Karim A, Muhammad T, Ahmad F. Lipid-Lowering Medications are Associated with Reduced Sarcopenia-Related Quality of Life in Older Adults with Hyperlipidemia. Drugs Aging 2024; 41:443-453. [PMID: 38564165 DOI: 10.1007/s40266-024-01111-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Statins medications negatively affect age-associated loss of muscle mass and strength, termed sarcopenia, and neuromuscular junction (NMJ) integrity. However, their association with the sarcopenia-related-quality-of-life (SarQoL) is unknown. METHODS In this cross-sectional, case control study, we recruited male nonusers (n = 75 and age 75.2 ± 5.9 years) and users (n = 77 and age 77.1 ± 6.2 years) of statins to evaluate SarQoL and handgrip strength (HGS). We also measured plasma C-terminal agrin fragment-22 (CAF22) as a marker of NMJ degradation. RESULTS Statin users had higher CAF22, and lower HGS, and cumulative SarQoL scores than non-users (all p < 0.05). Plasma CAF22 exhibited negative correlations with SarQoL scores for physical and mental health, locomotion, functionality, activities-of-daily-living, and cumulative SarQoL in statins users and non-users (all p < 0.05). Lastly, the cumulative SarQoL scores exhibited positive associations with HGS and gait speed in the study participants (all p < 0.05). CONCLUSIONS Collectively, statin usage was associated with NMJ degradation and reduced SarQoL. Statins should be cautiously prescribed in patients with sarcopenia with reduced QoL.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences, M27-122, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates.
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| | - Imran M Khan
- Burn and Plastic Surgery Center, Hayatabad Medical Complex, Hayatabad, Peshawar, Pakistan
| | - Asima Karim
- Department of Basic Medical Sciences, M27-122, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Iron Biology Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Tahir Muhammad
- Department of Biochemistry, Gomal Medical College, Dera Ismail Khan, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, M27-122, College of Medicine, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
11
|
Meng H, Liao Z, Ji Y, Wang D, Han Y, Huang C, Hu X, Chen J, Zhang H, Li Z, Wang C, Sun H, Sun J, Chen L, Yin J, Zhao J, Xu T, Liu H. FGF7 enhances the expression of ACE2 in human islet organoids aggravating SARS-CoV-2 infection. Signal Transduct Target Ther 2024; 9:104. [PMID: 38654010 PMCID: PMC11039711 DOI: 10.1038/s41392-024-01790-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/04/2024] [Accepted: 03/10/2024] [Indexed: 04/25/2024] Open
Abstract
The angiotensin-converting enzyme 2 (ACE2) is a primary cell surface viral binding receptor for SARS-CoV-2, so finding new regulatory molecules to modulate ACE2 expression levels is a promising strategy against COVID-19. In the current study, we utilized islet organoids derived from human embryonic stem cells (hESCs), animal models and COVID-19 patients to discover that fibroblast growth factor 7 (FGF7) enhances ACE2 expression within the islets, facilitating SARS-CoV-2 infection and resulting in impaired insulin secretion. Using hESC-derived islet organoids, we demonstrated that FGF7 interacts with FGF receptor 2 (FGFR2) and FGFR1 to upregulate ACE2 expression predominantly in β cells. This upregulation increases both insulin secretion and susceptibility of β cells to SARS-CoV-2 infection. Inhibiting FGFR counteracts the FGF7-induced ACE2 upregulation, subsequently reducing viral infection and replication in the islets. Furthermore, retrospective clinical data revealed that diabetic patients with severe COVID-19 symptoms exhibited elevated serum FGF7 levels compared to those with mild symptoms. Finally, animal experiments indicated that SARS-CoV-2 infection increased pancreatic FGF7 levels, resulting in a reduction of insulin concentrations in situ. Taken together, our research offers a potential regulatory strategy for ACE2 by controlling FGF7, thereby protecting islets from SARS-CoV-2 infection and preventing the progression of diabetes in the context of COVID-19.
Collapse
Affiliation(s)
- Hao Meng
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511495, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Zhiying Liao
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511495, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Yanting Ji
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Dong Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yang Han
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, 430023, Hubei, China
| | - Chaolin Huang
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, 430023, Hubei, China
| | - Xujuan Hu
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan, 430023, Hubei, China
| | - Jingyi Chen
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Hengrui Zhang
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Zonghong Li
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Changliang Wang
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Hui Sun
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Jiaqi Sun
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Lihua Chen
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Jiaxiang Yin
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China
| | - Jincun Zhao
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China.
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Tao Xu
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511495, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China.
| | - Huisheng Liu
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511495, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou, 510320, Guangdong, China.
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
12
|
Gómez-Moyano E, Pavón-Morón J, Rodríguez-Capitán J, Bardán-Rebollar D, Ramos-Carrera T, Villalobos-Sánchez A, Pérez de Pedro I, Ruiz-García FJ, Mora-Robles J, López-Sampalo A, Pérez-Velasco MA, Bernal-López MR, Gómez-Huelgas R, Jiménez-Navarro M, Romero-Cuevas M, Costa F, Trenas A, Pérez-Belmonte LM. The Role of Heparin in Postural Orthostatic Tachycardia Syndrome and Other Post-Acute Sequelae of COVID-19. J Clin Med 2024; 13:2405. [PMID: 38673677 PMCID: PMC11050777 DOI: 10.3390/jcm13082405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
The therapeutic management and short-term consequences of the coronavirus disease 2019 (COVID-19) are well known. However, COVID-19 post-acute sequelae are less known and represent a public health problem worldwide. Patients with COVID-19 who present post-acute sequelae may display immune dysregulation, a procoagulant state, and persistent microvascular endotheliopathy that could trigger microvascular thrombosis. These elements have also been implicated in the physiopathology of postural orthostatic tachycardia syndrome, a frequent sequela in post-COVID-19 patients. These mechanisms, directly associated with post-acute sequelae, might determine the thrombotic consequences of COVID-19 and the need for early anticoagulation therapy. In this context, heparin has several potential benefits, including immunomodulatory, anticoagulant, antiviral, pro-endothelial, and vascular effects, that could be helpful in the treatment of COVID-19 post-acute sequelae. In this article, we review the evidence surrounding the post-acute sequelae of COVID-19 and the potential benefits of the use of heparin, with a special focus on the treatment of postural orthostatic tachycardia syndrome.
Collapse
Affiliation(s)
- Elisabeth Gómez-Moyano
- Servicio de Dermatología, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
| | - Javier Pavón-Morón
- Servicio de Cardiología, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (J.P.-M.); (M.J.-N.); (M.R.-C.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), IBIMA-Plataforma BIONAND, Universidad de Málaga (UMA), 29010 Málaga, Spain;
| | - Jorge Rodríguez-Capitán
- Servicio de Cardiología, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (J.P.-M.); (M.J.-N.); (M.R.-C.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), IBIMA-Plataforma BIONAND, Universidad de Málaga (UMA), 29010 Málaga, Spain;
| | | | | | - Aurora Villalobos-Sánchez
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain (I.P.d.P.); (A.L.-S.); (M.-R.B.-L.); (R.G.-H.)
| | - Iván Pérez de Pedro
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain (I.P.d.P.); (A.L.-S.); (M.-R.B.-L.); (R.G.-H.)
| | | | - Javier Mora-Robles
- Servicio de Cardiología, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Almudena López-Sampalo
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain (I.P.d.P.); (A.L.-S.); (M.-R.B.-L.); (R.G.-H.)
| | - Miguel A. Pérez-Velasco
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain (I.P.d.P.); (A.L.-S.); (M.-R.B.-L.); (R.G.-H.)
| | - Maria-Rosa Bernal-López
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain (I.P.d.P.); (A.L.-S.); (M.-R.B.-L.); (R.G.-H.)
- Centro de Investigación en Red Fisiopatología de la Obesidad y la Nutrtición (CIBERObn), IBIMA-Plataforma BIONAND, Universidad de Málaga (UMA), 29010 Málaga, Spain
| | - Ricardo Gómez-Huelgas
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain (I.P.d.P.); (A.L.-S.); (M.-R.B.-L.); (R.G.-H.)
- Centro de Investigación en Red Fisiopatología de la Obesidad y la Nutrtición (CIBERObn), IBIMA-Plataforma BIONAND, Universidad de Málaga (UMA), 29010 Málaga, Spain
| | - Manuel Jiménez-Navarro
- Servicio de Cardiología, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (J.P.-M.); (M.J.-N.); (M.R.-C.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), IBIMA-Plataforma BIONAND, Universidad de Málaga (UMA), 29010 Málaga, Spain;
| | - Miguel Romero-Cuevas
- Servicio de Cardiología, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; (J.P.-M.); (M.J.-N.); (M.R.-C.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), IBIMA-Plataforma BIONAND, Universidad de Málaga (UMA), 29010 Málaga, Spain;
| | - Francesco Costa
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, A.O.U. Policlinic ‘G. Martino’, Via C. Valeria 1, 98165 Messina, Italy;
| | - Alicia Trenas
- Servicio de Medicina Interna, Área Sanitaria Norte de Málaga, Hospital de Antequera, 29200 Antequera, Spain;
| | - Luis M. Pérez-Belmonte
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), IBIMA-Plataforma BIONAND, Universidad de Málaga (UMA), 29010 Málaga, Spain;
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain (I.P.d.P.); (A.L.-S.); (M.-R.B.-L.); (R.G.-H.)
- Servicio de Medicina Interna, Hospital Helicópteros Sanitarios, 29660 Marbella, Spain
| |
Collapse
|
13
|
Khan S, Ahmad F, Khalid N. Applications of Strain-Specific Probiotics in the Management of Cardiovascular Diseases: A Systemic Review. Mol Nutr Food Res 2024; 68:e2300675. [PMID: 38549453 DOI: 10.1002/mnfr.202300675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/14/2024] [Indexed: 05/08/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of global mortality and novel approaches for prevention and management are needed. The human gastrointestinal tract hosts a diverse microbiota that is crucial in maintaining metabolic homeostasis. The formulation of effective probiotics, alone or in combination, has been under discussion due to their impact on cardiovascular and metabolic diseases. Probiotics have been shown to impact cardiovascular health positively. An imbalance in the presence of Firmicutes and Bacteroidetes has been linked to the progression of CVDs due to their impact on bile acid and cholesterol metabolism. The probiotics primarily help in the reduction of plasma low-density lipoprotein levels and attenuation of the proinflammatory markers. These beneficial microorganisms contribute to lowering cholesterol levels and produce essential short-chain fatty acids. The impact of lipid-regulating probiotic strains on human health is quite significant. However, only a few have been tested for potential beneficial efficacy, and ambiguity exists regarding strain dosages, interactions with confounding factors, and potential adverse effects. Hence, more comprehensive studies and randomized trials are needed to understand the mechanisms of probiotics on CVDs and to ensure human health. This review assesses the evidence and highlights the roles of strain-specific probiotics in the management of CVDs.
Collapse
Affiliation(s)
- Saleha Khan
- Department of Human Nutrition and Dietetics, School of Food and Agricultural Sciences, University of Management and Technology, Lahore, 54000, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Nauman Khalid
- Department of Human Nutrition and Dietetics, School of Food and Agricultural Sciences, University of Management and Technology, Lahore, 54000, Pakistan
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, 59911, United Arab Emirates
| |
Collapse
|
14
|
Qaisar R, Iqbal MS, Karim A, Muhammad T, Ahmad F. A leaky gut contributes to reduced sarcopenia-related quality of life (SarQoL) in geriatric older adults. Qual Life Res 2024; 33:551-559. [PMID: 37930557 DOI: 10.1007/s11136-023-03547-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2023] [Indexed: 11/07/2023]
Abstract
PURPOSE The sarcopenia quality-of-life (SarQoL) questionnaire is designed to evaluate the quality of life of sarcopenic patients. A pathological increase in intestinal permeability leads to several systemic diseases, but its contribution to SarQoL is unknown. METHODS We recruited controls (n = 84, age = 74.6 ± 4.9 years) and sarcopenic (n = 55, age = 76.1 ± 4.2 years) men for validating and adapting a Pashto version of SarQoL. We measured the scores for seven domains of SarQoL, body composition, and handgrip strength (HGS). We also measured plasma zonulin as a marker of increased intestinal permeability. RESULTS The Pashto SarQoL exhibited adequate discriminative ability, construct validity, internal consistency, and test-retest reliability, without exhibiting the floor and ceiling effect. Sarcopenic patients had higher plasma zonulin and lower scores on SarQoL domains for physical and mental health, locomotion, body composition, functionality, activities of daily living, leisure, and fear, and cumulative SarQoL scores than controls. Plasma zonulin exhibited significant coefficients of determination with Pashto SarQoL domains for locomotion (r2 = 0.217), functionality (r2 = 0.101), activities of daily living (r2 = 0.302), and cumulative SarQoL scores (r2 = 0.168). We also found high efficacies of zonulin in diagnosing low scores for functionality (AUC = 0.785, 95% C.I = 0.708-0.863), activities of daily living (AUC = 0.785, 95% C.I = 0.708-0.863), and cumulative SarQoL scores (AUC = 0.821, 95% C.I = 0.751-0.891). CONCLUSION Altogether, SarQoL appears reliable in measuring the quality of life in sarcopenic patients. A leaky gut has a potential contribution to reduced SarQoL in sarcopenia.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| | - M Shahid Iqbal
- Department of Neurology and Stroke Medicine, Rehman Medical Institute, Peshawar, 25124, Pakistan
| | - Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Tahir Muhammad
- Department of Biochemistry, Gomal Medical College, Dera Ismail Khan, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Space Medicine Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
15
|
Moll-Bernardes R, Ferreira JR, Sousa AS, Tortelly MB, Pimentel AL, Figueiredo ACBS, Schaustz EB, Secco JCP, Sales ARK, Terzi FVO, Xavier de Brito A, Sarmento RO, Noya-Rabelo MM, Fortier S, Matos E Silva FA, Vera N, Conde L, Cabral-Castro MJ, Albuquerque DC, Rosado de-Castro P, Camargo GC, Pinheiro MVT, Souza OF, Bozza FA, Luiz RR, Medei E. Impact of the immune profiles of hypertensive patients with and without obesity on COVID-19 severity. Int J Obes (Lond) 2024; 48:254-262. [PMID: 37932408 DOI: 10.1038/s41366-023-01407-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Comorbidities such as obesity, hypertension, and diabetes are associated with COVID-19 development and severity, probably due to immune dysregulation; however, the mechanisms underlying these associations are not clear. The immune signatures of hypertensive patients with obesity with COVID-19 may provide new insight into the mechanisms of immune dysregulation and progression to severe disease in these patients. METHODS Hypertensive patients were selected prospectively from a multicenter registry of adults hospitalized with COVID-19 and stratified according to obesity (BMI ≥ 30 kg/m²). Clinical data including baseline characteristics, complications, treatment, and 46 immune markers were compared between groups. Logistic regression was performed to identify variables associated with the risk of COVID-19 progression in each group. RESULTS The sample comprised 213 patients (89 with and 124 without obesity). The clinical profiles of patients with and without obesity differed, suggesting potential interactions with COVID-19 severity. Relative to patients without obesity, patients with obesity were younger and fewer had cardiac disease and myocardial injury. Patients with obesity had higher EGF, GCSF, GMCSF, interleukin (IL)-1ra, IL-5, IL-7, IL-8, IL-15, IL-1β, MCP 1, and VEGF levels, total lymphocyte counts, and CD8+ CD38+ mean fluorescence intensity (MFI), and lower NK-NKG2A MFI and percentage of CD8+ CD38+ T cells. Significant correlations between cytokine and immune cell expression were observed in both groups. Five variables best predicted progression to severe COVID-19 in patients with obesity: diabetes, the EGF, IL-10, and IL-13 levels, and the percentage of CD8+ HLA-DR+ CD38+ cells. Three variables were predictive for patients without obesity: myocardial injury and the percentages of B lymphocytes and HLA-DR+ CD38+ cells. CONCLUSION Our findings suggest that clinical and immune variables and obesity interact synergistically to increase the COVID-19 progression risk. The immune signatures of hypertensive patients with and without obesity severe COVID-19 highlight differences in immune dysregulation mechanisms, with potential therapeutic applications.
Collapse
Affiliation(s)
| | - Juliana R Ferreira
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology and Internal Medicine Department, Rede D'Or São Luiz, São Paulo, Brazil
| | - Andréa Silvestre Sousa
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Evandro Chagas National Institute of Infectious Disease, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Mariana B Tortelly
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology and Internal Medicine Department, Rede D'Or São Luiz, São Paulo, Brazil
| | - Adriana L Pimentel
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology and Internal Medicine Department, Rede D'Or São Luiz, São Paulo, Brazil
| | - Ana Cristina B S Figueiredo
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology and Internal Medicine Department, Rede D'Or São Luiz, São Paulo, Brazil
| | | | | | | | - Flavia V O Terzi
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology and Internal Medicine Department, Rede D'Or São Luiz, São Paulo, Brazil
| | | | - Renée O Sarmento
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Internal Medicine Department, Rio de Janeiro Federal State University, Rio de Janeiro, Brazil
| | - Marcia M Noya-Rabelo
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology and Internal Medicine Department, Rede D'Or São Luiz, São Paulo, Brazil
- Bahia School of Medicine and Public Health, Bahia, Brazil
| | - Sergio Fortier
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | | | - Narendra Vera
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luciana Conde
- Bahia School of Medicine and Public Health, Bahia, Brazil
| | - Mauro Jorge Cabral-Castro
- Institute of Microbiology Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
- Department of Pathology, Faculty of Medicine, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Denilson C Albuquerque
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology Department, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | | | | | - Olga F Souza
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Cardiology and Internal Medicine Department, Rede D'Or São Luiz, São Paulo, Brazil
| | - Fernando A Bozza
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Evandro Chagas National Institute of Infectious Disease, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ronir R Luiz
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Institute for Studies in Public Health-IESC, UFRJ, Rio de Janeiro, Brazil
| | - Emiliano Medei
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.
- National Center for Structural Biology and Bioimaging, UFRJ, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Duijvelaar E, Gisby J, Peters JE, Bogaard HJ, Aman J. Longitudinal plasma proteomics reveals biomarkers of alveolar-capillary barrier disruption in critically ill COVID-19 patients. Nat Commun 2024; 15:744. [PMID: 38272877 PMCID: PMC10811341 DOI: 10.1038/s41467-024-44986-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
The pathobiology of respiratory failure in COVID-19 consists of a complex interplay between viral cytopathic effects and a dysregulated host immune response. In critically ill patients, imatinib treatment demonstrated potential for reducing invasive ventilation duration and mortality. Here, we perform longitudinal profiling of 6385 plasma proteins in 318 hospitalised patients to investigate the biological processes involved in critical COVID-19, and assess the effects of imatinib treatment. Nine proteins measured at hospital admission accurately predict critical illness development. Next to dysregulation of inflammation, critical illness is characterised by pathways involving cellular adhesion, extracellular matrix turnover and tissue remodelling. Imatinib treatment attenuates protein perturbations associated with inflammation and extracellular matrix turnover. These proteomic alterations are contextualised using external pulmonary RNA-sequencing data of deceased COVID-19 patients and imatinib-treated Syrian hamsters. Together, we show that alveolar capillary barrier disruption in critical COVID-19 is reflected in the plasma proteome, and is attenuated with imatinib treatment. This study comprises a secondary analysis of both clinical data and plasma samples derived from a clinical trial that was registered with the EU Clinical Trials Register (EudraCT 2020-001236-10, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001236-10/NL ) and Netherlands Trial Register (NL8491, https://www.trialregister.nl/trial/8491 ).
Collapse
Affiliation(s)
- Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| | - Jack Gisby
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - James E Peters
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, UK
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Pociūtė A, Kriaučiūnaitė K, Kaušylė A, Zablockienė B, Alčauskas T, Jelinskaitė A, Rudėnaitė A, Jančorienė L, Ročka S, Verkhratsky A, Pivoriūnas A. Plasma of COVID-19 Patients Does Not Alter Electrical Resistance of Human Endothelial Blood-Brain Barrier In Vitro. FUNCTION 2024; 5:zqae002. [PMID: 38486975 PMCID: PMC10935481 DOI: 10.1093/function/zqae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 03/17/2024] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 instigated the most serious global health crisis. Clinical presentation of COVID-19 frequently includes severe neurological and neuropsychiatric symptoms. However, it is presently unknown whether and to which extent pathological impairment of blood-brain barrier (BBB) contributes to the development of neuropathology during COVID-19 progression. In the present study, we used human induced pluripotent stem cells-derived brain endothelial cells (iBECs) to study the effects of blood plasma derived from COVID-19 patients on the BBB integrity in vitro. We also performed a comprehensive analysis of the cytokine and chemokine profiles in the plasma of COVID-19 patients, healthy and recovered individuals. We found significantly increased levels of interferon γ-induced protein 10 kDa, hepatocyte growth factor, and interleukin-18 in the plasma of COVID-19 patients. However, blood plasma from COVID-19 patients did not affect transendothelial electrical resistance in iBEC monolayers. Our results demonstrate that COVID-19-associated blood plasma inflammatory factors do not affect BBB paracellular pathway directly and suggest that pathological remodeling (if any) of BBB during COVID-19 may occur through indirect or yet unknown mechanisms.
Collapse
Affiliation(s)
- Agnė Pociūtė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Karolina Kriaučiūnaitė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Aida Kaušylė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| | - Birutė Zablockienė
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Tadas Alčauskas
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
| | - Augustė Jelinskaitė
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Akvilė Rudėnaitė
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Ligita Jančorienė
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
- Centre of Infectious Diseases, Vilnius University Hospital Santaros Klinikos, LT-08406 Vilnius, Lithuania
| | - Saulius Ročka
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
- Center of Neurosurgery, Vilnius University Hospital Santaros Klinikos, LT-08661 Vilnius, Lithuania
| | - Alexei Verkhratsky
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, 110052, China
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania
| |
Collapse
|
18
|
Wolszczak-Biedrzycka B, Dorf J, Wojewódzka-Żelezniakowicz M, Żendzian-Piotrowska M, Dymicka-Piekarska V, Matowicka-Karna J, Maciejczyk M. Changes in chemokine and growth factor levels may be useful biomarkers for monitoring disease severity in COVID-19 patients; a pilot study. Front Immunol 2024; 14:1320362. [PMID: 38239363 PMCID: PMC10794366 DOI: 10.3389/fimmu.2023.1320362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Aim The aim of the present study was to assess differences in the serum levels of chemokines and growth factors (GFs) between COVID-19 patients and healthy controls. The diagnostic utility of the analyzed proteins for monitoring the severity of the SARS-CoV- 2 infection based on the patients' MEWS scores was also assessed. Materials and methods The serum levels of chemokines and growth factors were analyzed in hospitalized COVID-19 patients (50 women, 50 men) with the use of the Bio-Plex Pro™ Human Cytokine Screening Panel (Biorad) and the Bio-Plex Multiplex system. Results The study demonstrated that serum levels of MIP-1α, RANTES, Eotaxin, CTACK, GRO-α, IP-10, MIG, basic-FGF, HGF, SCGF-β, G-CSF, M-CSF, SCF, MIF, LIF, and TRAIL were significant higher in COVID-19 patients than in the control group. The concentrations of CTACK, GRO-α, IP-10, MIG, basic-FGF, HGF, PDGF- BB, GM-CSF, SCF, LIF, and TRAIL were higher in asymptomatic/mildly symptomatic COVID-19 patients (stage 1) and COVID-19 patients with pneumonia without respiratory failure (stage 2). The receiver operating characteristic (ROC) analysis revealed that IP-10, MIF, MIG, and basic-FGF differentiated patients with COVID-19 from healthy controls with the highest sensitivity and specificity, whereas GM-CSF, basic-FGF, and MIG differentiated asymptomatic/mildly symptomatic COVID-19 patients (stage 1) from COVID-19 patients with pneumonia without respiratory failure (stage 2) with the highest sensitivity and specificity. Conclusions MIG, basic-FGF, and GM-CSF can be useful biomarkers for monitoring disease severity in patients with COVID-19.
Collapse
Affiliation(s)
- Blanka Wolszczak-Biedrzycka
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Justyna Dorf
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | | | | | | | - Joanna Matowicka-Karna
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
19
|
Gysan MR, Milacek C, Bal C, Zech A, Brugger J, Milos RI, Antoniewicz L, Idzko M, Gompelmann D. Ventilatory support and inflammatory peptides in hospitalised patients with COVID-19: A prospective cohort trial. PLoS One 2023; 18:e0293532. [PMID: 37917760 PMCID: PMC10621867 DOI: 10.1371/journal.pone.0293532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/14/2023] [Indexed: 11/04/2023] Open
Abstract
PURPOSE Several studies have shown that SARS-CoV-2 can induce a massive release of cytokines which contributes to disease severity and mortality. Therefore, cytokine levels in the serum may help to predict disease severity and survival in COVID-19 patients. METHODS In this prospective trial, 88 patients who were hospitalised for COVID-19 were enrolled. Blood samples for serum peptide measurements were taken at the time closest to hospitalisation, at day 5, 9 and 13 (±1). The concentrations of cytokines (IL-1α, IL-1β, IL-1RA, IL-6, L-7, L-10, IFN-γ and TNF-α), chemokines (CCL-3, CCL-4 and CCL-7) and growth factors (G-CSF, GM-CSF and VEGF) were assessed and correlated with the type of ventilation, occurrence of consolidations on imaging and the level of care. RESULTS COVID-19 patients (median age 68 years, IQR 55-77) stayed in hospital between 5-171 days. Compared to patients in the general care unit, patients in the intermediate care unit (IMCU) and intensive care unit (ICU) presented significantly elevated serum IL-6 (p = 0.004) and lower IFN-γ levels (p = 0.005), respectively. The peak inspiratory pressure in ventilated patients correlated positively with IL-1RA, G-CSF and inversely with IFN-γ serum levels (all p<0.05). VEGF serum levels inversely correlated with the fraction of inspired oxygen in patients receiving high-flow nasal canula oxygen therapy (p = 0.047). No significant correlation between serum concentrations of the measured peptides and the type of ventilation, occurrence of radiological consolidations or in-hospital mortality has been observed. CONCLUSION IL1-RA, IL-6, IFN-γ, G-CSF, CCL-7 and VEGF serum levels could prove helpful as biomarkers to assess disease severity and the need for intensive care in COVID-19 patients.
Collapse
Affiliation(s)
- Maximilian Robert Gysan
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christopher Milacek
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christina Bal
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Andreas Zech
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Jonas Brugger
- Institute for Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Ruxandra-Iulia Milos
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukasz Antoniewicz
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Daniela Gompelmann
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Bacharaki D, Karagiannis M, Giannakopoulos P, Papachristou E, Divanis D, Sardeli A, Petrou D, Nikolopoulos P, Bratsiakou A, Zoi V, Piliouras N, Damoraki G, Liakopoulos V, Goumenos D, Giamarellos-Bourboulis EJ. Immune responses of patients on maintenance hemodialysis after infection by SARS-CoV-2: a prospective observational cohort study. BMC Infect Dis 2023; 23:581. [PMID: 37674148 PMCID: PMC10481459 DOI: 10.1186/s12879-023-08569-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Immune dysregulation in patients with acute COVID-19 under chronic hemodialysis (CHD) is fully not elucidated. The changes of mononuclear counts and mediators before and after HD and associations with final outcome were studied. METHOD In this prospective study, hospitalized patients with moderate-to-severe COVID-19 under CHD and matched comparators under HD were analyzed for their absolute counts of lymphoid cells and circulating inflammatory mediators. Blood samples were collected before start and at the end of the first HD session; dialysate samples were also collected. RESULT Fifty-nine patients with acute COVID-19 under CHD and 20 uninfected comparators under CHD were enrolled. Circulating concentrations of tumor necrosis factor-alpha (TNFα), interleukin (IL)-10, interferon-γ and platelet-derived growth factor-A were increased in patients. Concentrations of mediators did not differ before and after HD. Significant decreases of CD4-lymphocytes and CD19-lymphocytes were found in patients. The decrease of the expression of HLA-DR on CD14-monocytes was associated with unfavorable outcome (defined as WHO-CPS 6 or more by day 28); increased counts of CD19-lymphocytes were associated with better outcomes. CONCLUSION Patients under CHD develop an inflammatory reaction to SARS-CoV-2 characterized by increase of inflammatory mediators, decrease of circulating T-lymphocytes and decrease of the expression of HLA-DR on CD14-monocytes.
Collapse
Affiliation(s)
- Dimitra Bacharaki
- Department of Nephrology, University General Hospital Attikon, Athens, Greece.
| | - Minas Karagiannis
- Department of Nephrology, University General Hospital Attikon, Athens, Greece
| | | | - Evangelos Papachristou
- Department of Nephrology, Rion University Hospital, University of Patras, Patras, Greece
| | - Dimitrios Divanis
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aggeliki Sardeli
- Department of Nephrology, University General Hospital Attikon, Athens, Greece
| | - Dimitra Petrou
- Department of Nephrology, University General Hospital Attikon, Athens, Greece
| | - Petros Nikolopoulos
- Department of Nephrology, University General Hospital Attikon, Athens, Greece
| | - Adamantia Bratsiakou
- Department of Nephrology, Rion University Hospital, University of Patras, Patras, Greece
| | - Vassiliki Zoi
- Department of Nephrology, University General Hospital Attikon, Athens, Greece
| | - Nikitas Piliouras
- Department of Nephrology, University General Hospital Attikon, Athens, Greece
| | - Georgia Damoraki
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilios Liakopoulos
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios Goumenos
- Department of Nephrology, Rion University Hospital, University of Patras, Patras, Greece
| | | |
Collapse
|
21
|
Qaisar R, Karim A, Iqbal MS, Ahmad F, Shaikh A, Kamli H, Khamjan NA. A leaky gut contributes to postural dysfunction in patients with Alzheimer's disease. Heliyon 2023; 9:e19485. [PMID: 37662779 PMCID: PMC10472051 DOI: 10.1016/j.heliyon.2023.e19485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/19/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023] Open
Abstract
Background Postural dysfunction is a common problem in patients with Alzheimer's disease (AD) and may lead to functional dependency and increasing morbidity and mortality. However, the pathophysiology of postural dysfunction in AD patients remains poorly understood. Objectives Elevated intestinal permeability is an underlying contributor to multiple diseases, including AD. We aimed to investigate the association of elevated intestinal permeability with postural dysfunction in AD patients. Design Setting Participants Measurements We conducted a cross-sectional, observational study on older adults, including controls and AD patients. We investigated the associations of postural balance with plasma zonulin, a marker of elevated intestinal permeability in geriatric controls (n = 74) and patients with mild (n = 71) and moderate (n = 66) AD. We used a standardized physical performance battery to measure balance in supine, tandem, and semi-tandem positions. We also measured handgrip strength (HGS), and gait speed as markers of physical capacity. Results AD patients exhibited lower balance scores, HGS, and gait speed and higher plasma zonulin than in controls (all p < 0.05). Plasma zonulin levels demonstrated significant areas under the curves in diagnosing poor balance in AD patients (all p < 0.05). Moderate AD was associated with lower balance and physical capacity, and higher zonulin than mild AD (ALL P < 0.05). Poor scores on balance scale were associated with higher expressions of markers of inflammation, oxidative stress, and muscle damage providing a mechanistic link between increased intestinal permeability and postural dysfunction in AD patients. Conclusion The results of our study show that plasma zonulin measurement may be used to diagnose postural dysfunction in AD patients. The study is relevant to non-ambulant and/or comatose AD patients with postural dysfunction. Our findings also highlight the therapeutic potential of repairing the intestinal leak to improve postural control and reduce the risk of falls in AD patients.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - M. Shahid Iqbal
- Department of Neurology and Stroke Medicine, Rehman Medical Institute, Peshawar, Pakistan
| | - Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, 59911, United Arab Emirates
| | - Ahmad Shaikh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia
| | - Hossam Kamli
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia
| | - Nizar A. Khamjan
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, 45142, Saudi Arabia
| |
Collapse
|
22
|
Ahmad F, Karim A, Khan J, Qaisar R. Circulating H-FABP as a biomarker of frailty in patients with chronic heart failure. Exp Biol Med (Maywood) 2023; 248:1383-1392. [PMID: 37787063 PMCID: PMC10657591 DOI: 10.1177/15353702231198080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/10/2023] [Indexed: 10/04/2023] Open
Abstract
Increased vulnerability to physiologic stressors, termed frailty, is a common occurrence in patients with chronic heart failure (CHF). However, the definite biomarkers to assess frailty in CHF patients are not known. Here, we assessed the frailty phenotype and its potential association with heart failure (HF) markers in CHF patients. We categorized controls (n = 59) and CHF patients (n = 80), the participants, into robust, pre-frail, and frail based on the cardiovascular health study (CHS) frailty index. The plasma levels of HF markers, including tumorigenicity 2 (s-ST2), galectin-3, and heart-type fatty acid binding protein (H-FABP), were measured and correlated with frailty phenotype and cardiac function. The levels of plasma s-ST2, galectin-3, and H-FABP were profoundly elevated in CHF patients. Conversely, the frailty index scores were significantly lower in ischemic and non-ischemic CHF patients versus controls. Of the assessed HF markers, only H-FABP was positively correlated (r2 = 0.07, P = 0.02) with the frailty score in CHF patients. Collectively, these observations suggest that circulating H-FABP may serve as a biomarker of frailty in CHF patients.
Collapse
Affiliation(s)
- Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Javaidullah Khan
- Department of Cardiology, Post Graduate Medical Institute, Hayatabad Medical Complex, Peshawar 25120, Pakistan
| | - Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
23
|
Krenytska D, Strubchevska K, Kozyk M, Vovk T, Halenova T, Kot L, Raksha N, Savchuk O, Falalyeyeva T, Tsyryuk O, Ostapchenko L. Circulating levels of inflammatory cytokines and angiogenesis-related growth factors in patients with osteoarthritis after COVID-19. Front Med (Lausanne) 2023; 10:1168487. [PMID: 37484856 PMCID: PMC10358362 DOI: 10.3389/fmed.2023.1168487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/25/2023] [Indexed: 07/25/2023] Open
Abstract
Background The disease COVID-19, caused by SARS-CoV-2 infection, has a systemic effect and is associated with a number of pathophysiological mechanisms that mobilize a wide range of biomolecules. Cytokines and growth factors (GFs) are critical regulators of tissue damage or repair in osteoarthritis (OA) and are being recognized as key players in the pathogenesis of COVID-19. A clear understanding of the long-term consequences of SARS-CoV-2 infection, especially in patients with concomitant chronic diseases, is limited and needs to be elucidated. The study aimed to evaluate the degree of inflammation and levels of pro-angiogenic and hypoxic factors, as well as heat shock proteins HSP60 and HSP70 in plasma, of patients with OA after recovery from COVID-19. Methods The research involved patients of an orthopedic specialty clinic aged 39 to 80 diagnosed with knee OA. All examined patients were divided into three groups: the Control group included conditionally healthy donors, group OA included patients with knee OA mainly stage II or III and the group of OA and COVID-19 included patients with OA who had COVID-19. The plasma levels of pro-inflammatory molecules IL-1β, IL-6, TNF-α, NF-κB, angiogenic factors VEGF, FGF-2, PDGF, hypoxic factor HIF-1α and molecular chaperones HSP60 and HSP70 were measured by enzyme-linked immunosorbent assay. Results The study showed that in both groups of patients, with OA and convalescent COVID-19, there was an increase in the plasma level of IL-1β and a decrease in TNF-α and NF-κB levels when compared to healthy controls. Systemic deregulation of the cytokine profile was accompanied by reduction in plasma levels of pro-angiogenic growth factors, most pronounced in cases of VEGF and PDGF. This analysis did not reveal any significant difference in the plasma level of HIF-1α. A decrease in the level of stress protein HSP60 in the blood of patients with OA, as well as those patients who have had SARS-CoV-2 infection, has been established. Conclusion The results suggest the potential role pro-inflammatory cytokines and angiogenesis-related growth factors in pathogenesis of both joint pathologies and long-term systemic post-COVID-19 disorders.
Collapse
Affiliation(s)
- Daryna Krenytska
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | | - Marko Kozyk
- William Beaumont Hospital, Royal Oak, MI, United States
| | - Tetiana Vovk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetiana Halenova
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Larysa Kot
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Nataliia Raksha
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olexii Savchuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olena Tsyryuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Liudmyla Ostapchenko
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| |
Collapse
|
24
|
Qaisar R, Karim A, Iqbal MS, Alkahtani SA, Ahmad F, Kamli H. ACE Inhibitors Improve Skeletal Muscle by Preserving Neuromuscular Junctions in Patients with Alzheimer's Disease. J Alzheimers Dis 2023:JAD230201. [PMID: 37334602 DOI: 10.3233/jad-230201] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Hypertension and skeletal muscle decline are common findings in patients with Alzheimer's disease (AD). Angiotensin-converting enzyme (ACE) inhibitors preserve skeletal muscle and physical capacity; however, the driving mechanisms are poorly understood. OBJECTIVE We investigated the effects of ACE inhibitors on the neuromuscular junction (NMJ) with relevance to skeletal muscle and physical capacity in AD patients and age-matched controls. METHODS We evaluated controls (n = 59) and three groups of AD patients, including normotensive (n = 51) and patients with hypertension taking ACE inhibitors (n = 53) or other anti-hypertensive medications (n = 49) at baseline and one year later. We measure plasma c-terminal agrin fragment-22 (CAF22) as a marker of NMJ degradation, handgrip strength (HGS), and Short Physical Performance Battery (SPPB) as markers of physical capacity. RESULTS At baseline AD patients demonstrated lower HGS and SPPB scores and higher CAF22 levels than controls, irrespective of the hypertension status (all p < 0.05). The use of ACE inhibitors was associated with higher HGS and relative maintenance of SPPB scores, gait speed, and plasma CAF22 levels. Conversely, other anti-hypertensive medications were associated with an unaltered HGS, reduced SPPB scores and elevated plasma CAF22 levels (both p < 0.05). We also found dynamic associations of CAF22 with HGS, gait speed, and SPPB in AD patients taking ACE inhibitors (all p < 0.05). These changes were associated with reduced oxidative stress in AD patients taking ACE inhibitors (p < 0.05). CONCLUSION Altogether, ACE inhibitors are associated with higher HGS, preserved physical capacity, and the prevention of NMJ degradation in hypertensive AD patients.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - M Shahid Iqbal
- Department of Neurology and Stroke Medicine, Rehman Medical Institute, Peshawar, Pakistan
| | - Shaea A Alkahtani
- Exercise Physiology Department, College of Sport Sciences and Physical Activity, King Saud University, Riyadh, Saudi Arabia
| | - Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Hossam Kamli
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
25
|
Petersen E, Chudakova D, Erdyneeva D, Zorigt D, Shabalina E, Gudkov D, Karalkin P, Reshetov I, Mynbaev OA. COVID-19-The Shift of Homeostasis into Oncopathology or Chronic Fibrosis in Terms of Female Reproductive System Involvement. Int J Mol Sci 2023; 24:ijms24108579. [PMID: 37239926 DOI: 10.3390/ijms24108579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 coronavirus remains a global public health concern due to the systemic nature of the infection and its long-term consequences, many of which remain to be elucidated. SARS-CoV-2 targets endothelial cells and blood vessels, altering the tissue microenvironment, its secretion, immune-cell subpopulations, the extracellular matrix, and the molecular composition and mechanical properties. The female reproductive system has high regenerative potential, but can accumulate damage, including due to SARS-CoV-2. COVID-19 is profibrotic and can change the tissue microenvironment toward an oncogenic niche. This makes COVID-19 and its consequences one of the potential regulators of a homeostasis shift toward oncopathology and fibrosis in the tissues of the female reproductive system. We are looking at SARS-CoV-2-induced changes at all levels in the female reproductive system.
Collapse
Affiliation(s)
- Elena Petersen
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Daria Chudakova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Daiana Erdyneeva
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Dulamsuren Zorigt
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | | | - Denis Gudkov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Pavel Karalkin
- P.A. Herzen Moscow Research Institute of Oncology, 125284 Moscow, Russia
- Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Igor Reshetov
- Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ospan A Mynbaev
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| |
Collapse
|
26
|
Vassiliou AG, Vrettou CS, Keskinidou C, Dimopoulou I, Kotanidou A, Orfanos SE. Endotheliopathy in Acute COVID-19 and Long COVID. Int J Mol Sci 2023; 24:8237. [PMID: 37175942 PMCID: PMC10179170 DOI: 10.3390/ijms24098237] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
The pulmonary endothelium is a highly regulated organ that performs a wide range of functions under physiological and pathological conditions. Since endothelial dysfunction has been demonstrated to play a direct role in sepsis and acute respiratory distress syndrome, its role in COVID-19 has also been extensively investigated. Indeed, apart from the COVID-19-associated coagulopathy biomarkers, new biomarkers were recognised early during the pandemic, including markers of endothelial cell activation or injury. We systematically searched the literature up to 10 March 2023 for studies examining the association between acute and long COVID-19 severity and outcomes and endothelial biomarkers.
Collapse
Affiliation(s)
- Alice G. Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| | | | | | | | | | - Stylianos E. Orfanos
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (C.K.); (I.D.); (A.K.)
| |
Collapse
|
27
|
Ahmad F, Karim A, Khan J, Qaisar R. Plasma Galectin-3 and H-FABP correlate with poor physical performance in patients with congestive heart failure. Exp Biol Med (Maywood) 2023; 248:532-540. [PMID: 36803120 PMCID: PMC10281532 DOI: 10.1177/15353702231151980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/26/2022] [Indexed: 02/22/2023] Open
Abstract
Heart failure (HF) is often associated with compromised physical capacity in patients. However, it is unclear if established HF markers correlate with the physical performance of patients with congestive HF (CHF). We assessed the left ventricular end-systolic dimension (LVESD) and ejection fraction (LVEF) and, physical performance parameters, including short physical performance battery (SPPB), gait speed (GS), and handgrip strength (HGS) in 80 patients with CHF along with 59 healthy controls. Furthermore, levels of plasma HF markers galectin-3 and heart-specific fatty acid binding protein (H-FABP) were measured in relation to the severity of HF and physical performance. Irrespective of etiology, significantly greater LVESD and lower LVEF were observed in HF patients versus controls. As expected, the levels of HF markers galectin-3 and H-FABP were upregulated in the CHF patients which were accompanied by significantly elevated levels of plasma zonulin and inflammatory marker C-reactive protein (CRP). The SPPB scores, GS, and HGS were significantly lower in the ischemic and non-ischemic HF patients than controls. The level of galectin-3 was inversely correlated with SPPB scores (r2 = 0.089, P = 0.01) and HGS (r2 = 0.078, P = 0.01). Similarly, H-FABP levels were also inversely correlated with SPPB scores (r2 = 0.06, P = 0.03) and HGS (r2 = 0.109, P = 0.004) in the patients with CHF. Taken together, CHF adversely affects physical performance, and galectin-3 and H-FABP may serve as biomarkers of physical disability in patients with CHF. The robust correlations of galectin-3 and H-FABP with the physical performance parameters and CRP in CHF patients suggest that the poor physical performance may partly be caused due to systemic inflammation.
Collapse
Affiliation(s)
- Firdos Ahmad
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, 59911 United Arab Emirates
| | - Asima Karim
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Javaidullah Khan
- Department of Cardiology, Post Graduate Medical Institute, Hayatabad Medical Complex, Peshawar 25120, Pakistan
| | - Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
28
|
Miguel-Pastor L, Satué K, Chicharro D, Peláez P, Torres-Torrillas M, Carrillo JM, Cerón JJ, Sopena JJ, Rubio M. Evaluation of Platelet-Rich Plasma by means of PRGF ®-Endoret ® protocol in leukemia cats: PDGF-BB and TGF-ß1 valuation. Front Vet Sci 2023; 10:1110055. [PMID: 36777664 PMCID: PMC9909748 DOI: 10.3389/fvets.2023.1110055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction Feline leukemia virus (FeLV) is a chronic disease that leads to the weakening of a cat's immune system. Platelet-rich plasma (PRP) offers therapeutic effects for multiple diseases, the use of PRP and growth factors (GFs) determination could be an alternative treatment to improve the quality of life in these patients. The objectives of this study were to determine and compare the concentration of platelets (PLTs), red blood cells (RBCs) and white blood cells (WBCs) between samples of whole blood (WB), PRP and platelet-poor plasma (PPP) fractions, and to evaluate the concentration of platelet-derived growth factor BB (PDGF-BB) and transforming growth factor β1 (TGF-β1) in both fractions in FeLV cats using a PRGF®-Endoret® protocol previously standardized in this species. Methods WB was collected from 11 asymptomatic FeLV-positive cats. PRP and PPP was obtained following PRGF®-Endoret® technology according to centrifugation at 265 g for 10 min. Cellular components, RBCs, WBCs, PLTs, and the PDGF-BB and TGF-β1 concentrations in PRP and PPP fractions were determined. Results PLT in the PRP fraction was statistically higher than WB and PPP fraction, with no statistical differences between WB and PPP. PLT concentration increased 1.4 times in PRP fraction compared to WB. Mean platelet volume (MPV) did not differ significantly between the WB, PRP, and PPP fractions. Compared to WB, the absolute numbers of RBCs and WBCs were decreased by 99% and more than 95% in the PRP and PPP fractions, respectively. TGF-ß1 concentrations increased in PRP vs. PPP, with no changes in PDGF-BB. Discussion Based on the degree of PLT enrichment and the absence of RBCs and WBCs, this blood product could be classified as a Pure Platelet-Rich Plasma (P-PRP). The presence of GFs in PRP and PPP samples suggests that the PRGF®-Endoret® methodology is suitable for obtaining PRP in FeLV cats, despite future studies are necessary to optimize the technique, standardize the results and assess clinical efficacy.
Collapse
Affiliation(s)
- Laura Miguel-Pastor
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Katy Satué
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - José M. Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain,García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - José J. Cerón
- Interdisciplinary Laboratory of Clinical Analysis, University of Murcia, Murcia, Spain
| | - Joaquín J. Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain,García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain,*Correspondence: Joaquín J. Sopena ✉
| | - Mónica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain,García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| |
Collapse
|
29
|
Papadopoulos KI, Papadopoulou A, Aw TC. A protective erythropoietin evolutionary landscape, NLRP3 inflammasome regulation, and multisystem inflammatory syndrome in children. Hum Cell 2023; 36:26-40. [PMID: 36310304 PMCID: PMC9618415 DOI: 10.1007/s13577-022-00819-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2022]
Abstract
The low incidence of pediatric severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection and the associated multisystem inflammatory syndrome (MIS-C) lack a unifying pathophysiological explanation, impeding effective prevention and therapy. Activation of the NACHT, LRR, and PYD domains-containing protein (NLRP) 3 inflammasome in SARS-CoV-2 with perturbed regulation in MIS-C, has been reported. We posit that, early age physiological states and genetic determinants, such as certain polymorphisms of renin-angiotensin aldosterone system (RAAS) molecules, promote a controlled RAAS hyperactive state, and form an evolutionary landscape involving an age-dependent erythropoietin (EPO) elevation, mediating ancestral innate immune defenses that, through appropriate NLRP3 regulation, mitigate tissue injury and pathogen invasion. SARS-CoV-2-induced downregulation of angiotensin-converting enzyme (ACE)2 expression in endothelial cells (EC), impairment of endothelial nitric oxide (NO) synthase (eNOS) activity and downstream NO bioavailability, may promote a hyperactive RAAS with elevated angiotensin II and aldosterone that, can trigger, and accelerate NLRP3 inflammasome activation, while EPO-eNOS/NO abrogate it. Young age and a protective EPO evolutionary landscape may successfully inhibit SARS-CoV-2 and contain NLRP3 inflammasome activation. By contrast, increasing age and falling EPO levels, in genetically susceptible children with adverse genetic variants and co-morbidities, may lead to unopposed RAAS hyperactivity, NLRP3 inflammasome dysregulation, severe endotheliitis with pyroptotic cytokine storm, and development of autoantibodies, as already described in MIS-C. Our haplotype estimates, predicted from allele frequencies in population databases, are in concordance with MIS-C incidence reports in Europeans but indicate lower risks for Asians and African Americans. Targeted Mendelian approaches dissecting the influence of relevant genetic variants are needed.
Collapse
Affiliation(s)
- Konstantinos I Papadopoulos
- Department of Research and Development, THAI StemLife Co., Ltd., 566/3 THAI StemLife Bldg., Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd., Wangthonglang, 10310, Bangkok, Thailand.
| | - Alexandra Papadopoulou
- Occupational and Environmental Health Services, Feelgood Lund, Ideon Science Park, Scheelevägen 17, 223 63, Lund, Sweden
| | - Tar-Choon Aw
- Department of Laboratory Medicine, Changi General Hospital, Singapore, 529889, Singapore
- Department of Medicine, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|