1
|
Ahmed G, Abdelgadir Y, Abdelghani A, Simpson P, Barbeau J, Basel D, Barrios CS, Smith BA, Schilter KF, Udani R, Reddi HV, Willoughby RE. Reduction in ACE2 expression in peripheral blood mononuclear cells during COVID-19 - implications for post COVID-19 conditions. BMC Infect Dis 2024; 24:663. [PMID: 38956476 PMCID: PMC11221185 DOI: 10.1186/s12879-024-09321-0] [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: 09/20/2023] [Accepted: 04/14/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Severe COVID-19 is uncommon, restricted to 19% of the total population. In response to the first virus wave (alpha variant of SARS-CoV-2), we investigated whether a biomarker indicated severity of disease and, in particular, if variable expression of angiotensin converting enzyme 2 (ACE2) in blood might clarify this difference in risk and of post COVID -19 conditions (PCC). METHODS The IRB-approved study compared patients hospitalized with severe COVID-19 to healthy controls. Severe infection was defined requiring oxygen or increased oxygen need from baseline at admission with positive COVID-19 PCR. A single blood sample was obtained from patients within a day of admission. ACE2 RNA expression in blood cells was measured by an RT-PCR assay. Plasma ACE1 and ACE2 enzyme activities were quantified by fluorescent peptides. Plasma TIMP-1, PIIINP and MMP-9 antigens were quantified by ELISA. Data were entered into REDCap and analyzed using STATA v 14 and GraphPad Prism v 10. RESULTS Forty-eight patients and 72 healthy controls were recruited during the pandemic. ACE2 RNA expression in peripheral blood mononuclear cells (PBMC) was rarely detected acutely during severe COVID-19 but common in controls (OR for undetected ACE2: 12.4 [95% CI: 2.62-76.1]). ACE2 RNA expression in PBMC did not determine plasma ACE1 and ACE2 activity, suggesting alternative cell-signaling pathways. Markers of fibrosis (TIMP-1 and PIIINP) and vasculopathy (MMP-9) were additionally elevated. ACE2 RNA expression during severe COVID-19 often responded within hours to convalescent plasma. Analogous to oncogenesis, we speculate that potent, persistent, cryptic processes following COVID-19 (the renin-angiotensin system (RAS), fibrosis and vasculopathy) initiate or promote post-COVID-19 conditions (PCC) in susceptible individuals. CONCLUSIONS This work elucidates biological and temporal plausibility for ACE2, TIMP1, PIIINP and MMP-9 in the pathogenesis of PCC. Intersection of these independent systems is uncommon and may in part explain the rarity of PCC.
Collapse
Affiliation(s)
- Gulrayz Ahmed
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | - Pippa Simpson
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jody Barbeau
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Donald Basel
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | | - Rupa Udani
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Honey V Reddi
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Rodney E Willoughby
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
- Pediatric Infectious Diseases, C450, Medical College of Wisconsin, PO Box 1997, Milwaukee, WI 53201-1997, USA.
| |
Collapse
|
2
|
Aldarondo DA, Huynh C, Dickey L, Bilynsky C, Lee Y, Wayne EC. Nanoparticle endocytosis is driven by monocyte phenotype rather than nanoparticle size under high shear flow conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547038. [PMID: 37425838 PMCID: PMC10327044 DOI: 10.1101/2023.06.29.547038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Monocytes are members of the mononuclear phagocyte system involved in pathogen clearance and nanoparticle pharmacokinetics. Monocytes play a critical role in the development and progression of cardiovascular disease and, recently, in SARS-CoV-2 pathogenesis. While studies have investigated the effect of nanoparticle modulation on monocyte uptake, their capacity for nanoparticle clearance is poorly studied. In this study, we investigated the impact of ACE2 deficiency, frequently observed in individuals with cardiovascular complications, on monocyte nanoparticle endocytosis. Moreover, we investigated nanoparticle uptake as a function of nanoparticle size, physiological shear stress, and monocyte phenotype. Our Design of Experiment (DOE) analysis found that the THP-1 ACE2 - cells showed a greater preference for 100nm particles under atherosclerotic conditions than THP-1 wild-type cells. Observing how nanoparticles can modulate monocytes in the context of disease can inform precision dosing.
Collapse
|
3
|
Bueno V, Destro PH, Teixeira D, Frasca D. Angiotensin Converting Enzyme 1 Expression in the Leukocytes of Adults Aged 64 to 67 Years. JMIRX MED 2023; 4:e45220. [PMID: 37725526 PMCID: PMC10414256 DOI: 10.2196/45220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 09/21/2023]
Abstract
The renin angiotensin system is composed of several enzymes and substrates on which angiotensin converting enzyme (ACE) 1 and renin act to produce angiotensin II. ACE1 and its substrates control blood pressure, affect cardiovascular and renal function, hematopoiesis, reproduction, and immunity. The increased expression of ACE1 has been observed in human monocytes during congestive heart failure and abdominal aortic aneurysm. Moreover, T lymphocytes from individuals with hypertension presented increased expression of ACE1 after in vitro stimulation with angiotensin II (ATII) with the highest ACE1 expression observed in individuals with hypertension with low-grade inflammation. Our group and others have shown that aging is associated with comorbidities, chronic inflammation, and immunosenescence, but there is a lack of data about ACE1 expression on immune cells during the aging process. Therefore, our aim was to evaluate the levels of ACE1 expression in nonlymphoid cells compared to lymphoid that in cells in association with the immunosenescence profile in adults older than 60 years. Cryopreserved peripheral blood mononuclear cells obtained from blood samples were used. Cells were stained with monoclonal antibodies and evaluated via flow cytometry. We found that ACE1 was expressed in 56.9% of nonlymphocytes and in more than 90% of lymphocytes (all phenotypes). All donors exhibited characteristics of immunosenescence, as evaluated by low frequencies of naïve CD4+ and CD8+ T cells, high frequencies of effector memory re-expressing CD45RA CD8+ T cells, and double-negative memory B cells. These findings, in addition to the increased C-reactive protein levels, are intriguing questions for the study of ACE1, inflammaging, immunosenescence, and perspectives for drug development or repurposing (Reviewed by the Plan P #PeerRef Community).
Collapse
Affiliation(s)
- Valquiria Bueno
- Division of Immunology, Department of Microbiology Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Pedro Henrique Destro
- Division of Immunology, Department of Microbiology Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela Teixeira
- Division of Immunology, Department of Microbiology Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela Frasca
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
4
|
Kamthe DD, Sarangkar SD, Dalvi MS, Gosavi NA, Nikam VS. Angiotensin converting enzyme 2 level and its significance in COVID-19 and other diseases patients. Eur J Clin Invest 2023; 53:e13891. [PMID: 36222740 PMCID: PMC9874405 DOI: 10.1111/eci.13891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Angiotensin-converting enzyme 2 (ACE2) expressions and its modulation are of great interest as being a key receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) and the protective arm of the rennin-angiotensin axis, maintaining cardiovascular homeostasis. However, ACE2 expressions and their modulation in the healthy and disease background are yet to be explored. METHOD We performed a meta-analysis, extracting the data for ACE2 expression in human subjects with various diseases, including SARS-CoV2 infection without or with co-morbidity. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. Out of 203 studies, 39 met the inclusion criteria with SARS-CoV2 patients without co-morbidity, SARS-CoV2 patients with co-morbidity, cardiovascular (CVD) patients, diabetes patients, kidney disorders patients, pulmonary disease patients, and other viral infections patients. RESULTS Angiotensin-converting enzyme 2 expression was significantly increased in all diseases. There was an elevated level of ACE2, especially membrane-bound ACE2, in COVID-19 patients compared to healthy controls. A statistically significant increase in ACE2 expression was observed in CVD patients and patients with other viral diseases compared to healthy subjects. Moreover, subgroup analysis of ACE2 expression as soluble and membrane-bound ACE2 revealed a remarkable increase in membrane-bound ACE2 in CVD patients, patients with viral infection compared to soluble ACE2 and pooled standard mean difference (SMD) with the random-effects model was 0.37 and 2.23 respectively. CONCLUSION It was observed that utilizing the ACE2 by SARS-CoV2 for its entry and its consequence leads to several complications. So there is a need to investigate the underlying mechanism along with novel therapeutic strategies.
Collapse
Affiliation(s)
- Dipanjali Dhananjay Kamthe
- Department of Pharmacology, STES's, Smt. Kashibai Navale College of PharmacySavitribai Phule Pune UniversityPuneIndia
| | - Swapnil Dilip Sarangkar
- Department of Pharmacology, STES's, Smt. Kashibai Navale College of PharmacySavitribai Phule Pune UniversityPuneIndia
| | - Manali Suresh Dalvi
- Department of Pharmacology, STES's, Smt. Kashibai Navale College of PharmacySavitribai Phule Pune UniversityPuneIndia
| | - Netra Arun Gosavi
- Department of Pharmacology, STES's, Smt. Kashibai Navale College of PharmacySavitribai Phule Pune UniversityPuneIndia
| | - Vandana Sandeep Nikam
- Department of Pharmacology, STES's, Smt. Kashibai Navale College of PharmacySavitribai Phule Pune UniversityPuneIndia
| |
Collapse
|
5
|
Ait-Belkacem I, Cartagena García C, Millet-Wallisky E, Izquierdo N, Loosveld M, Arnoux I, Morange PE, Galland F, Lambert N, Malergue F, Busnel JM. SARS-CoV-2 spike protein induces a differential monocyte activation that may contribute to age bias in COVID-19 severity. Sci Rep 2022; 12:20824. [PMID: 36460710 PMCID: PMC9716544 DOI: 10.1038/s41598-022-25259-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
A strong bias related to age is observed in COVID-19 patients with pediatric subjects developing a milder disease than adults. We hypothesized that a specific SARS-CoV-2 effect conjugated with preexisting differences in the immune systems may explain this. Using flow cytometry, we investigated basal immune differences in a cohort consisting of 16 non-infected young and 16 aged individuals and further leveraged an in vitro whole blood model of SARS-CoV-2 infection so that functional differences could be mined as well. In short, blood diluted in culture media was incubated 5 or 24 h with the trimeric spike protein or controls. Following unsupervised analysis, we first confirmed that the immune lymphoid and myeloid systems in adults are less efficient and prone to develop higher inflammation than those in children. We notably identified in adults a higher CD43 lymphocyte expression, known for its potentially inhibitory role. The spike protein induced different responses between adults and children, notably a higher increase of inflammatory markers together with lower monocyte and B cell activation in adults. Interestingly, CD169, a CD43 ligand overexpressed in COVID-19 patients, was confirmed to be strongly modulated by the spike protein. In conclusion, the spike protein exacerbated the preexisting lower immune responsiveness and higher inflammatory potential in adults. Altogether, some of the markers identified may explain the marked age bias and be predictive of severity.
Collapse
Affiliation(s)
- Ines Ait-Belkacem
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
- Aix Marseille Université CNRS INSERM CIML Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Celia Cartagena García
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
- INSERM UMRs 1097, Aix Marseille University, Marseille, France
| | - Ewa Millet-Wallisky
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
| | - Nicolas Izquierdo
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
| | - Marie Loosveld
- Hematology Laboratory, Timone University Hospital, APHM, Marseille, France
| | - Isabelle Arnoux
- Hematology Laboratory, Timone University Hospital, APHM, Marseille, France
| | | | - Franck Galland
- Aix Marseille Université CNRS INSERM CIML Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | | | - Fabrice Malergue
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France.
| | - Jean-Marc Busnel
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France.
| |
Collapse
|
6
|
Mitra J, Kodavati M, Provasek VE, Rao KS, Mitra S, Hamilton DJ, Horner PJ, Vahidy FS, Britz GW, Kent TA, Hegde ML. SARS-CoV-2 and the central nervous system: Emerging insights into hemorrhage-associated neurological consequences and therapeutic considerations. Ageing Res Rev 2022; 80:101687. [PMID: 35843590 PMCID: PMC9288264 DOI: 10.1016/j.arr.2022.101687] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/20/2022] [Accepted: 07/07/2022] [Indexed: 01/27/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to impact our lives by causing widespread illness and death and poses a threat due to the possibility of emerging strains. SARS-CoV-2 targets angiotensin-converting enzyme 2 (ACE2) before entering vital organs of the body, including the brain. Studies have shown systemic inflammation, cellular senescence, and viral toxicity-mediated multi-organ failure occur during infectious periods. However, prognostic investigations suggest that both acute and long-term neurological complications, including predisposition to irreversible neurodegenerative diseases, can be a serious concern for COVID-19 survivors, especially the elderly population. As emerging studies reveal sites of SARS-CoV-2 infection in different parts of the brain, potential causes of chronic lesions including cerebral and deep-brain microbleeds and the likelihood of developing stroke-like pathologies increases, with critical long-term consequences, particularly for individuals with neuropathological and/or age-associated comorbid conditions. Our recent studies linking the blood degradation products to genome instability, leading to cellular senescence and ferroptosis, raise the possibility of similar neurovascular events as a result of SARS-CoV-2 infection. In this review, we discuss the neuropathological consequences of SARS-CoV-2 infection in COVID survivors, focusing on possible hemorrhagic damage in brain cells, its association to aging, and the future directions in developing mechanism-guided therapeutic strategies.
Collapse
Affiliation(s)
- Joy Mitra
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA.
| | - Manohar Kodavati
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Vincent E Provasek
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA; College of Medicine, Texas A&M University, College Station, TX, USA
| | - K S Rao
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation Deemed to be University, Green Fields, Vaddeswaram, Andhra Pradesh 522502, India
| | - Sankar Mitra
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Dale J Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA; Weill Cornell Medical College, New York, USA
| | - Philip J Horner
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA; Weill Cornell Medical College, New York, USA
| | - Farhaan S Vahidy
- Center for Outcomes Research, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Gavin W Britz
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA; Weill Cornell Medical College, New York, USA
| | - Thomas A Kent
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, USA
| | - Muralidhar L Hegde
- Division of DNA Repair Research, Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, USA; Weill Cornell Medical College, New York, USA.
| |
Collapse
|
7
|
Jha NK, Ojha S, Jha SK, Dureja H, Singh SK, Shukla SD, Chellappan DK, Gupta G, Bhardwaj S, Kumar N, Jeyaraman M, Jain R, Muthu S, Kar R, Kumar D, Goswami VK, Ruokolainen J, Kesari KK, Singh SK, Dua K. Evidence of Coronavirus (CoV) Pathogenesis and Emerging Pathogen SARS-CoV-2 in the Nervous System: A Review on Neurological Impairments and Manifestations. J Mol Neurosci 2021; 71:2192-2209. [PMID: 33464535 PMCID: PMC7814864 DOI: 10.1007/s12031-020-01767-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is an issue of global significance that has taken the lives of many across the world. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for its pathogenesis. The pulmonary manifestations of COVID-19 have been well described in the literature. Initially, it was thought to be limited to the respiratory system; however, we now recognize that COVID-19 also affects several other organs, including the nervous system. Two similar human coronaviruses (CoV) that cause severe acute respiratory syndrome (SARS-CoV-1) and Middle East respiratory syndrome (MERS-CoV) are also known to cause disease in the nervous system. The neurological manifestations of SARS-CoV-2 infection are growing rapidly, as evidenced by several reports. There are several mechanisms responsible for such manifestations in the nervous system. For instance, post-infectious immune-mediated processes, direct virus infection of the central nervous system (CNS), and virus-induced hyperinflammatory and hypercoagulable states are commonly involved. Guillain-Barré syndrome (GBS) and its variants, dysfunction of taste and smell, and muscle injury are numerous examples of COVID-19 PNS (peripheral nervous system) disease. Likewise, hemorrhagic and ischemic stroke, encephalitis, meningitis, encephalopathy acute disseminated encephalomyelitis, endothelialitis, and venous sinus thrombosis are some instances of COVID-19 CNS disease. Due to multifactorial and complicated pathogenic mechanisms, COVID-19 poses a large-scale threat to the whole nervous system. A complete understanding of SARS-CoV-2 neurological impairments is still lacking, but our knowledge base is rapidly expanding. Therefore, we anticipate that this comprehensive review will provide valuable insights and facilitate the work of neuroscientists in unfolding different neurological dimensions of COVID-19 and other CoV associated abnormalities.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, 201310, UP, India.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, PO Box - 17666, United Arab Emirates University, Al Ain, UAE
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, 201310, UP, India
| | - Harish Dureja
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, Newcastle, NSW, 2305, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Shanu Bhardwaj
- Department of Biotechnology, HIMT, CCS University, Greater Noida, UP, India
| | - Neeraj Kumar
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Madhan Jeyaraman
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, UP, 201310, Greater Noida, India
| | - Rashmi Jain
- School of Medical Sciences and Research, Sharda University, UP, 201310, Greater Noida, India
| | - Sathish Muthu
- Research Associate, Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
| | - Rohan Kar
- Indian Institute of Management Ahmedabad (IIMA), Gujarat, 380015, India
| | - Dhruv Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India
| | - Vineet Kumar Goswami
- Department of Biological Sciences, School of Basic and Applied Sciences, G.D. Goenka University, G.D. Goenka Education City Sohna Gurugram Road, Haryana- 122103, India
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, 00076, Espoo, Finland
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, 00076, Espoo, Finland
| | - Sandeep Kumar Singh
- Centre of Biomedical Research, SGPGI Campus, Lucknow, 226014, UP, India
- Indian Scientific Education and Technology Foundation, Lucknow, 226002, UP, India
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, Newcastle, NSW, 2305, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Post box no. 9, Solan, Himachal Pradesh, 173229, India
| |
Collapse
|
8
|
Almengló C, Couselo-Seijas M, Agra RM, Varela-Román A, García-Acuña JM, González-Peteiro M, González-Juanatey JR, Eiras S, Álvarez E. Soluble angiotensin-converting enzyme levels in heart failure or acute coronary syndrome: revisiting its modulation and prognosis value. J Mol Med (Berl) 2021; 99:1741-1753. [PMID: 34529122 PMCID: PMC8443916 DOI: 10.1007/s00109-021-02129-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/21/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022]
Abstract
The main objective was to compare the meaning of soluble angiotensin-converting enzyme-2 (sACE2) plasma levels modulation on the prognosis of two cohorts of heart failure (HF) and acute coronary syndrome (ACS). We conducted an observational clinical study where sACE2 was measured in two cohorts of HF or ACS (102 patients each), matched by age and gender. The primary endpoint (cardiac death) and the secondary endpoints (non-fatal myocardial infarction or HF readmission) were registered during a 5-year follow-up period. Association with pharmacotherapy was studied, and the effects of cardiovascular drugs on ACE isoforms expression were analysed in human umbilical vein endothelial cells (HUVEC) in vitro. The levels of sACE2 were significantly higher in the HF than ACS cohort. sACE2 was inversely related with the leukocytes number and directly with urea levels. In the ACS cohort, sACE2 was associated with age and glycaemic parameters, but in the HF cohort, the association was with N-terminal pro-B-type natriuretic peptide. The levels of sACE2 were related to long-term prognosis and confirmed as a non-independent predictor in the HF cohort. Soluble ACE2 was higher in patients treated with angiotensin receptors blockers and β-blockers, accordingly with losartan and metoprolol upregulation of ACE1 and ACE2 in HUVECs. Plasma levels of sACE2 were higher in HF than in ACS, independently of age and gender, and were related to long-term cardiac death in the HF cohort. Losartan and metoprolol, but not enalapril, upregulated ACE expression in endothelial cells, accordingly with higher levels of sACE2 in patients using these drugs.
Collapse
Affiliation(s)
- Cristina Almengló
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain
| | - Marinela Couselo-Seijas
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain
| | - Rosa M Agra
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain.,CIBERCV, Madrid, Spain.,Servicio de Cardiología y Unidad de Hemodinámica, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain
| | - Alfonso Varela-Román
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain.,CIBERCV, Madrid, Spain.,Servicio de Cardiología y Unidad de Hemodinámica, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain
| | - José M García-Acuña
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain.,CIBERCV, Madrid, Spain.,Servicio de Cardiología y Unidad de Hemodinámica, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain
| | - Mercedes González-Peteiro
- Departamento de Enfermería, Universidad de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain
| | - José R González-Juanatey
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain.,CIBERCV, Madrid, Spain.,Servicio de Cardiología y Unidad de Hemodinámica, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain
| | - Sonia Eiras
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain.,CIBERCV, Madrid, Spain
| | - Ezequiel Álvarez
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana S/N, 15706, Santiago de Compostela, A Coruña, Spain. .,CIBERCV, Madrid, Spain. .,Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Universidad de Santiago de Compostela, 15782, Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
9
|
Tracz J, Luczak M. Applying Proteomics and Integrative "Omics" Strategies to Decipher the Chronic Kidney Disease-Related Atherosclerosis. Int J Mol Sci 2021; 22:7492. [PMID: 34299112 PMCID: PMC8305100 DOI: 10.3390/ijms22147492] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk of atherosclerosis and premature mortality, mainly due to cardiovascular events. However, well-known risk factors, which promote "classical" atherosclerosis are alone insufficient to explain the high prevalence of atherosclerosis-related to CKD (CKD-A). The complexity of the molecular mechanisms underlying the acceleration of CKD-A is still to be defied. To obtain a holistic picture of these changes, comprehensive proteomic approaches have been developed including global protein profiling followed by functional bioinformatics analyses of dysregulated pathways. Furthermore, proteomics surveys in combination with other "omics" techniques, i.e., transcriptomics and metabolomics as well as physiological assays provide a solid ground for interpretation of observed phenomena in the context of disease pathology. This review discusses the comprehensive application of various "omics" approaches, with emphasis on proteomics, to tackle the molecular mechanisms underlying CKD-A progression. We summarize here the recent findings derived from global proteomic approaches and underline the potential of utilizing integrative systems biology, to gain a deeper insight into the pathogenesis of CKD-A and other disorders.
Collapse
Affiliation(s)
| | - Magdalena Luczak
- European Centre for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland;
| |
Collapse
|
10
|
Semyachkina-Glushkovskaya O, Mamedova A, Vinnik V, Klimova M, Saranceva E, Ageev V, Yu T, Zhu D, Penzel T, Kurths J. Brain Mechanisms of COVID-19-Sleep Disorders. Int J Mol Sci 2021; 22:6917. [PMID: 34203143 PMCID: PMC8268116 DOI: 10.3390/ijms22136917] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
2020 and 2021 have been unprecedented years due to the rapid spread of the modified severe acute respiratory syndrome coronavirus around the world. The coronavirus disease 2019 (COVID-19) causes atypical infiltrated pneumonia with many neurological symptoms, and major sleep changes. The exposure of people to stress, such as social confinement and changes in daily routines, is accompanied by various sleep disturbances, known as 'coronasomnia' phenomenon. Sleep disorders induce neuroinflammation, which promotes the blood-brain barrier (BBB) disruption and entry of antigens and inflammatory factors into the brain. Here, we review findings and trends in sleep research in 2020-2021, demonstrating how COVID-19 and sleep disorders can induce BBB leakage via neuroinflammation, which might contribute to the 'coronasomnia' phenomenon. The new studies suggest that the control of sleep hygiene and quality should be incorporated into the rehabilitation of COVID-19 patients. We also discuss perspective strategies for the prevention of COVID-19-related BBB disorders. We demonstrate that sleep might be a novel biomarker of BBB leakage, and the analysis of sleep EEG patterns can be a breakthrough non-invasive technology for diagnosis of the COVID-19-caused BBB disruption.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Aysel Mamedova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Valeria Vinnik
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Maria Klimova
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Elena Saranceva
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Vasily Ageev
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
| | - Tingting Yu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dan Zhu
- Wuhan National Laboratory for Optoelectronics, Britton Chance Center for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China; (T.Y.); (D.Z.)
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Thomas Penzel
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Sleep Medicine Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Kurths
- Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany;
- Department of Biology, Saratov State University, Atrakhanskaya Str. 83, 410012 Saratov, Russia; (A.M.); (V.V.); (M.K.); (E.S.); (V.A.)
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
11
|
Jafari Khaljiri H, Jamalkhah M, Amini Harandi A, Pakdaman H, Moradi M, Mowla A. Comprehensive Review on Neuro-COVID-19 Pathophysiology and Clinical Consequences. Neurotox Res 2021; 39:1613-1629. [PMID: 34169404 PMCID: PMC8225460 DOI: 10.1007/s12640-021-00389-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/08/2023]
Abstract
Aside from the respiratory distress as the predominant clinical presentation of SARS-CoV-2 infection, various neurological complications have been reported with the infection during the ongoing pandemic, some of which cause serious morbidity and mortality. Herein, we gather the latest anatomical evidence of the virus's presence within the central nervous system. We then delve into the possible SARS-CoV-2 entry routes into the neurological tissues, with the hematogenous and the neuronal routes as the two utmost passage routes into the nervous system. We then give a comprehensive review of the neurological manifestations of the SARS-CoV-2 invasion in both the central and peripheral nervous system and its underlying pathophysiology via investigating large studies in the field and case reports in cases of study scarcity.
Collapse
Affiliation(s)
- Helia Jafari Khaljiri
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Monire Jamalkhah
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Ali Amini Harandi
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hossein Pakdaman
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Moradi
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashkan Mowla
- Division of Endovascular Neurosurgery, Department of Neurological Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA
| |
Collapse
|
12
|
Poznyak AV, Bharadwaj D, Prasad G, Grechko AV, Sazonova MA, Orekhov AN. Renin-Angiotensin System in Pathogenesis of Atherosclerosis and Treatment of CVD. Int J Mol Sci 2021; 22:ijms22136702. [PMID: 34206708 PMCID: PMC8269397 DOI: 10.3390/ijms22136702] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis has complex pathogenesis, which involves at least three serious aspects: inflammation, lipid metabolism alterations, and endothelial injury. There are no effective treatment options, as well as preventive measures for atherosclerosis. However, this disease has various severe complications, the most severe of which is cardiovascular disease (CVD). It is important to note, that CVD is among the leading causes of death worldwide. The renin–angiotensin–aldosterone system (RAAS) is an important part of inflammatory response regulation. This system contributes to the recruitment of inflammatory cells to the injured site and stimulates the production of various cytokines, such as IL-6, TNF-a, and COX-2. There is also an association between RAAS and oxidative stress, which is also an important player in atherogenesis. Angiotensin-II induces plaque formation at early stages, and this is one of the most crucial impacts on atherogenesis from the RAAS. Importantly, while stimulating the production of ROS, Angiotensin-II at the same time decreases the generation of NO. The endothelium is known as a major contributor to vascular function. Oxidative stress is the main trigger of endothelial dysfunction, and, once again, links RAAS to the pathogenesis of atherosclerosis. All these implications of RAAS in atherogenesis lead to an explicable conclusion that elements of RAAS can be promising targets for atherosclerosis treatment. In this review, we also summarize the data on treatment approaches involving cytokine targeting in CVD, which can contribute to a better understanding of atherogenesis and even its prevention.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Dwaipayan Bharadwaj
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110067, India;
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Gauri Prasad
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Margarita A. Sazonova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
- Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
13
|
Poznyak AV, Bezsonov EE, Eid AH, Popkova TV, Nedosugova LV, Starodubova AV, Orekhov AN. ACE2 Is an Adjacent Element of Atherosclerosis and COVID-19 Pathogenesis. Int J Mol Sci 2021; 22:ijms22094691. [PMID: 33946649 PMCID: PMC8124184 DOI: 10.3390/ijms22094691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/17/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a highly contagious new infection caused by the single-stranded RNA Sars-CoV-2 virus. For the first time, this infection was recorded in December 2019 in the Chinese province of Wuhan. The virus presumably crossed the interspecies barrier and passed to humans from a bat. Initially, the disease was considered exclusively in the context of damage to the respiratory system, but it quickly became clear that the disease also entails serious consequences from various systems, including the cardiovascular system. Among these consequences are myocarditis, myocardial damage, subsequent heart failure, myocardial infarction, and Takotsubo syndrome. On the other hand, clinical data indicate that the presence of chronic diseases in a patient aggravates the course and outcome of coronavirus infection. In this context, the relationship between COVID-19 and atherosclerosis, a condition preceding cardiovascular disease and other disorders of the heart and blood vessels, is particularly interesting. The renin-angiotensin system is essential for the pathogenesis of both coronavirus disease and atherosclerosis. In particular, it has been shown that ACE2, an angiotensin-converting enzyme 2, plays a key role in Sars-CoV-2 infection due to its receptor activity. It is noteworthy that this enzyme is important for the normal functioning of the cardiovascular system. Disruptions in its production and functioning can lead to various disorders, including atherosclerosis.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Evgeny E. Bezsonov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Tatyana V. Popkova
- V.A. Nasonova Institute of Rheumatology, 34A Kashirskoye Shosse, 115522 Moscow, Russia;
| | - Ludmila V. Nedosugova
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of the Russian Federation 8/2 Trubenskaya Street, 119991 Moscow, Russia;
| | - Antonina V. Starodubova
- Federal Research Centre for Nutrition, Biotechnology and Food Safety, 2/14 Ustinsky Passage, 109240 Moscow, Russia;
- Pirogov Russian National Research Medical University, 1 Ostrovitianov Street, 117997 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
14
|
Shehata GA, Lord KC, Grudzinski MC, Elsayed M, Abdelnaby R, Elshabrawy HA. Neurological Complications of COVID-19: Underlying Mechanisms and Management. Int J Mol Sci 2021; 22:4081. [PMID: 33920904 PMCID: PMC8071289 DOI: 10.3390/ijms22084081] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 is a severe respiratory disease caused by the newly identified human coronavirus (HCoV) Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The virus was discovered in December 2019, and in March 2020, the disease was declared a global pandemic by the World Health Organization (WHO) due to a high number of cases. Although SARS-CoV-2 primarily affects the respiratory system, several studies have reported neurological complications in COVID-19 patients. Headache, dizziness, loss of taste and smell, encephalitis, encephalopathy, and cerebrovascular diseases are the most common neurological complications that are associated with COVID-19. In addition, seizures, neuromuscular junctions' disorders, and Guillain-Barré syndrome were reported as complications of COVID-19, as well as neurodegenerative and demyelinating disorders. However, the management of these conditions remains a challenge. In this review, we discuss the prevalence, pathogenesis, and mechanisms of these neurological sequelae that are secondary to SARS-CoV-2 infection. We aim to update neurologists and healthcare workers on the possible neurological complications associated with COVID-19 and the management of these disease conditions.
Collapse
Affiliation(s)
- Ghaydaa A. Shehata
- Department of Neurology and Psychiatry, Assiut University Hospitals, Assiut 71511, Egypt;
| | - Kevin C. Lord
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA;
| | | | - Mohamed Elsayed
- Department of Psychiatry and Psychotherapy III, University of Ulm, Leimgrubenweg 12-14, 89075 Ulm, Germany;
| | - Ramy Abdelnaby
- Department of Neurology, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany;
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| |
Collapse
|
15
|
Neurological Sequelae in Patients with COVID-19: A Histopathological Perspective. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021. [PMID: 33546463 DOI: 10.3390/ijerph18041415.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Neuroinvasive properties of SARS-CoV-2 have allowed the hypothesis of several pathogenic mechanisms related to acute and chronic neurological sequelae. However, neuropathological correlates have been poorly systematically investigated, being retrieved from reports of single case or limited case series still. METHODS A PubMed search was carried out to review all publications on autopsy in subjects with "COronaVIrus Disease-19" (COVID-19). Among them, we focused on histological findings of the brain, which were compared with those from the authors' autoptic studies performed in some COVID-19 patients. RESULTS Only seven studies reported histological evidence of brain pathology in patients deceased for COVID-19, including three with reverse transcription-quantitative polymerase chain reaction evidence of viral infection. All these studies, in line with our experience, showed vascular-related and infection-related secondary inflammatory tissue damage due to an abnormal immune response. It is still unclear, however, whether these findings are the effect of a direct viral pathology or rather reflect a non-specific consequence of cardiovascular and pulmonary disease on the brain. CONCLUSIONS Notwithstanding the limited evidence available and the heterogeneity of the studies, we provide a preliminary description of the relationship between SARS-CoV-2 and brain sequelae. Systematic autoptic investigations are needed for accurate detection and adequate management of these patients.
Collapse
|
16
|
Fisicaro F, Di Napoli M, Liberto A, Fanella M, Di Stasio F, Pennisi M, Bella R, Lanza G, Mansueto G. Neurological Sequelae in Patients with COVID-19: A Histopathological Perspective. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041415. [PMID: 33546463 PMCID: PMC7913756 DOI: 10.3390/ijerph18041415] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/18/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neuroinvasive properties of SARS-CoV-2 have allowed the hypothesis of several pathogenic mechanisms related to acute and chronic neurological sequelae. However, neuropathological correlates have been poorly systematically investigated, being retrieved from reports of single case or limited case series still. METHODS A PubMed search was carried out to review all publications on autopsy in subjects with "COronaVIrus Disease-19" (COVID-19). Among them, we focused on histological findings of the brain, which were compared with those from the authors' autoptic studies performed in some COVID-19 patients. RESULTS Only seven studies reported histological evidence of brain pathology in patients deceased for COVID-19, including three with reverse transcription-quantitative polymerase chain reaction evidence of viral infection. All these studies, in line with our experience, showed vascular-related and infection-related secondary inflammatory tissue damage due to an abnormal immune response. It is still unclear, however, whether these findings are the effect of a direct viral pathology or rather reflect a non-specific consequence of cardiovascular and pulmonary disease on the brain. CONCLUSIONS Notwithstanding the limited evidence available and the heterogeneity of the studies, we provide a preliminary description of the relationship between SARS-CoV-2 and brain sequelae. Systematic autoptic investigations are needed for accurate detection and adequate management of these patients.
Collapse
Affiliation(s)
- Francesco Fisicaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (F.F.); (A.L.); (M.P.)
| | - Mario Di Napoli
- Department of Neurology and Stroke Unit, San Camillo de’ Lellis General Hospital, Viale Kennedy 1, 02100 Rieti, Italy; (M.D.N.); (M.F.)
| | - Aldo Liberto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (F.F.); (A.L.); (M.P.)
| | - Martina Fanella
- Department of Neurology and Stroke Unit, San Camillo de’ Lellis General Hospital, Viale Kennedy 1, 02100 Rieti, Italy; (M.D.N.); (M.F.)
| | - Flavio Di Stasio
- Department of Neurology and Stroke Unit Cesena-Forlì, Bufalini Hospital, AUSL Romagna, Viale Ghirotti 286, 47521 Cesena, Italy;
| | - Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (F.F.); (A.L.); (M.P.)
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia 87, 95123 Catania, Italy;
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgery Specialties, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
- Department of Neurology IC, Oasi Research Institute-IRCCS, Via Conte Ruggero 78, 94018 Troina, Italy
- Correspondence: ; Tel.: +39-095-3782448
| | - Gelsomina Mansueto
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia 2, 80138 Naples, Italy;
| |
Collapse
|
17
|
Salamanna F, Maglio M, Landini MP, Fini M. Body Localization of ACE-2: On the Trail of the Keyhole of SARS-CoV-2. Front Med (Lausanne) 2020; 7:594495. [PMID: 33344479 PMCID: PMC7744810 DOI: 10.3389/fmed.2020.594495] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023] Open
Abstract
The explosion of the new coronavirus (SARS-CoV-2) pandemic has brought the role of the angiotensin converting enzyme 2 (ACE2) back into the scientific limelight. Since SARS-CoV-2 must bind the ACE2 for entering the host cells in humans, its expression and body localization are critical to track the potential target organ of this infection and to outline disease progression and clinical outcomes. Here, we mapped the physiological body distribution, expression, and activities of ACE2 and discussed its potential correlations and mutal interactions with the disparate symptoms present in SARS-CoV-2 patients at the level of different organs. We highlighted that despite during SARS-CoV-2 infection ACE2-expressing organs may become direct targets, leading to severe pathological manifestations, and subsequent multiple organ failures, the exact mechanism and the potential interactions through which ACE2 acts in these organs is still heavily debated. Further scientific efforts, also considering a personalized approach aimed to consider specific patient differences in the mutual interactions ACE2-SARS-CoV-2 and the long-term health effects associated with COVID-19 are currently mandatory.
Collapse
Affiliation(s)
- Francesca Salamanna
- Surgical Sciences and Technologies, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Melania Maglio
- Surgical Sciences and Technologies, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Maria Paola Landini
- Scientific Direction, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Milena Fini
- Surgical Sciences and Technologies, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
18
|
Trojanowicz B, Ulrich C, Girndt M. Uremic Apelin and Leucocytic Angiotensin-Converting Enzyme 2 in CKD Patients. Toxins (Basel) 2020; 12:toxins12120742. [PMID: 33255902 PMCID: PMC7760850 DOI: 10.3390/toxins12120742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022] Open
Abstract
Apelin peptides (APLN) serve as second substrates for angiotensin-converting enzyme 2 (ACE2) and, in contrast to angiotensin II (AngII), exert blood-pressure lowering and vasodilatation effects through binding to G-coupled APLN receptor (APLNR). ACE2-mediated cleavage of the APLN may reduce its vasodilatory effects, but decreased ACE2 may potentiate the hypotensive properties of APLN. The role of APLN in uremia is unclear. We investigated the correlations between serum-APLN, leucocytic APLNR, and ACE2 in 32 healthy controls (NP), 66 HD, and 24 CKD3-5 patients, and the impact of APLN peptides on monocytic behavior and ACE2 expression under uremic conditions in vitro. We observed that serum APLN and leucocytic APLNR or SLCO2B1 were significantly elevated in uremic patients and correlated with decreased ACE2 on uremic leucocytes. APLN-treated THP-1 monocytes revealed significantly increased APLNR and ACE2, and reduced TNFa, IL-6, and MCSF. Uremic toxins induced a dramatic increase of miR-421 followed by significant reduction of ACE2 transcripts, partially counteracted with APLN-13 and -36. APLN-36 triggered the most potent transmigration and reduction of endothelial adhesion. These results suggest that although APLN peptides may partly protect against the decay of monocytic ACE2 transcripts, uremic milieu is the most dominant modulator of local ACE2, and likely to contribute to the progression of atherosclerosis.
Collapse
|
19
|
Pang L, Liu Y, Shen M, Ye J, Chen R, Lan Z, Wu Z, Guo Y, Zhang P. Influence of aging on deterioration of patients with COVID-19. Aging (Albany NY) 2020; 12:26248-26262. [PMID: 33232272 PMCID: PMC7803552 DOI: 10.18632/aging.202136] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
Aging is an important factor affecting the deterioration of patients with coronavirus disease 2019 (COVID-19). The aging and degeneration of various tissues and organs in the elderly lead to impaired organ function. Underlying conditions such as chronic lung disease, cardiovascular disease, and diabetes in aged patients are associated with higher mortality. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) primarily interacts with the cell surface receptor angiotensin-converting enzyme (ACE) 2 and other accessory proteins such as 78 kDa glucose-regulated protein 78 (GRP78) and CD147. Thus, altered receptor signals in aging and chronic disease play a role in SARS-CoV-2 infection, and are associated with a higher risk of deterioration in different organs. In this review, after a brief introduction to the link between aging and receptors for SARS-CoV-2, we focus on the risk of deterioration in different organs of COVID-19 patients considering aging as the main factor. We further discuss the structural and/or physiological changes in the immune system and organs (lung, heart, kidney, vessels, nerve system), as well as those associated with diabetes, in aging patients, and speculate on the most likely mechanisms underlying the deterioration of COVID-19 patients.
Collapse
Affiliation(s)
- Limin Pang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yi Liu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Maoze Shen
- Department of Internal Medicine, Raoping County People's Hospital, Chaozhou, Guangdong, People’s Republic of China
| | - Jujian Ye
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Ruirong Chen
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zhien Lan
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zhijian Wu
- Department of Cardiology, Boai Hospital of Zhongshan, Southern Medical University, Zhongshan, People’s Republic of China
| | - Yang Guo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Peidong Zhang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
20
|
Akki R, Fath N, Mohti H. COVID-19: Oxidative Preconditioning as a Potential Therapeutic Approach. ACS Chem Neurosci 2020; 11:3732-3740. [PMID: 33147964 PMCID: PMC7670822 DOI: 10.1021/acschemneuro.0c00453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
This Article summarizes the likely benefits of central nervous system oxidative preconditioning in the reduction of COVID-19 based on its putative pathogenesis. The current COVID-19 outbreak caused a pandemic with millions of infected patients and death cases worldwide. The clinical features of severe acute respiratory syndrome coronavirus (SARS-CoV) was initially linked with respiratory disorders, but recent studies have reported alterations of neurological and cerebrovascular functions in COVID-19 patients. The main viral infection features are related to cell death, inflammation, and cytokine generation, which can be associated with the dysregulation of redox systems or oxidative stress. However, until now, there is no available and effective therapeutic approach. Thus, it is necessary to search for care and adequate protection against the disease, especially for susceptible and vulnerable groups. Preconditioning, a well-known antioxidative stress and anti-inflammatory approach, is protective against many neurological age-related disorders. COVID-19 severity and morbidity have been observed in elderly patients. The aim of the present study is to elucidate the possible protective role of oxidative preconditioning in aged patients at high risk of developing severe COVID-19 complications.
Collapse
Affiliation(s)
- Rachid Akki
- Department
of Plant Protection, National School of
Agriculture-Meknes/ENA, BP S/40, Meknès 50001, Morocco
| | - Nada Fath
- Compared
Anatomy Unit, School of Veterinary Medicine, Hassan II Institute of Agronomy and Veterinary Medicine, Rabat 10000, Morocco
- Physiology
and Pathophysiology Laboratory, Department of Biology, Faculty of
Sciences, Mohamed V University, Rabat BP 8007.NU, Morocco
| | - Hicham Mohti
- Management
and Valorization of Natural Resources, Faculty of Sciences, Moulay Ismail University of Meknes, Meknes BP 11201, Morocco
| |
Collapse
|
21
|
Elberry MH, Ahmed H. Occult SARS-CoV-2 infection; a possible hypothesis for viral relapse. Med Hypotheses 2020; 144:109980. [PMID: 32570163 PMCID: PMC7833987 DOI: 10.1016/j.mehy.2020.109980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has emerged as a global public health emergency, which is characterized by high infection rate and fatal course. Recent data reported that the test for Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) RNA might become positive again after one or two consecutively negative tests. Many researchers are currently evaluating the clinical characteristics of the SARS-CoV-2 reactivation. In this letter, we proposed a possible mechanism of SARS-CoV-2 reactivation or relapse after negative nasopharyngeal swabs PCR.
Collapse
Affiliation(s)
- Mostafa H Elberry
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Egypt
| | - Hussien Ahmed
- Clinical Research Management Program, Edson College, Arizona State University, USA; Faculty of Medicine, Zagazig University, Sharkia, Egypt.
| |
Collapse
|
22
|
Wang F, Kream RM, Stefano GB. Long-Term Respiratory and Neurological Sequelae of COVID-19. Med Sci Monit 2020; 26:e928996. [PMID: 33177481 PMCID: PMC7643287 DOI: 10.12659/msm.928996] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022] Open
Abstract
Since the initial reports of coronavirus disease 2019 (COVID-19) in China in late 2019, infections from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have spread rapidly, resulting in a global pandemic that has caused millions of deaths. Initially, the large number of infected people required the direction of global healthcare resources to provide supportive care for the acutely ill population in an attempt to reduce mortality. While clinical trials for safe and effective antiviral agents are ongoing, and vaccine development programs are being accelerated, long-term sequelae of SARS-CoV-2 infection have become increasingly recognized and concerning. Although the upper and lower respiratory tracts are the main sites of entry of SARS-CoV-2 into the body, resulting in COVID-19 pneumonia as the most common presentation, acute lung damage may be followed by pulmonary fibrosis and chronic impairment of lung function, with impaired quality of life. Also, increasing reports have shown that SARS-CoV-2 infection involves the central nervous system (CNS) and the peripheral nervous system (PNS) and directly or indirectly damages neurons, leading to long-term neurological sequelae. This review aims to provide an update on the mechanisms involved in the development of the long-term sequelae of SARS-CoV-2 infection in the 3 main areas of lung injury, neuronal injury, and neurodegenerative diseases, including Alzheimer disease, Parkinson disease, and multiple sclerosis, and highlights the need for patient monitoring following the acute stage of infection with SARS-CoV-2 to provide a rationale for the prevention, diagnosis, and management of these potential long-term sequelae.
Collapse
Affiliation(s)
- Fuzhou Wang
- Group of Neuropharmacology and Neurophysiology, Division of Neuroscience, The Bonoi Academy of Science and Education, Chapel Hill, NC, U.S.A
| | | | - George B. Stefano
- International Scientific Information, Inc., Melville, NY, U.S.A
- Center for Cognitive and Molecular Neuroscience, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
23
|
Silva GM, França-Falcão MS, Calzerra NTM, Luz MS, Gadelha DDA, Balarini CM, Queiroz TM. Role of Renin-Angiotensin System Components in Atherosclerosis: Focus on Ang-II, ACE2, and Ang-1-7. Front Physiol 2020; 11:1067. [PMID: 33013457 PMCID: PMC7494970 DOI: 10.3389/fphys.2020.01067] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is the leading cause of vascular disease worldwide and contributes significantly to deaths from cardiovascular complications. There is a remarkably close relationship between atherosclerotic plaque formation and the activation of renin-angiotensin system (RAS). However, depending on which RAS pathway is activated, pro- or anti-atherogenic outcomes may be observed. This brief review focuses on the role of three of the most important pieces of RAS axis, angiotensin II (Ang-II), angiotensin converting enzyme type 2 (ACE2), and angiotensin 1-7 (Ang-1-7) and their involvement in atherosclerosis. We focused on the effects of these molecules on vascular function and inflammation, which are important determinants of atherogenesis. Furthermore, we highlighted potential pharmacological approaches to treat this disorder.
Collapse
Affiliation(s)
- Gabriela M Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | | | | | - Mickael S Luz
- Center of Biotechnology, Federal University of Paraiba, João Pessoa, Brazil
| | | | - Camille M Balarini
- Health Sciences Center, Federal University of Paraiba, João Pessoa, Brazil
| | - Thyago M Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| |
Collapse
|
24
|
Battaglini D, Brunetti I, Anania P, Fiaschi P, Zona G, Ball L, Giacobbe DR, Vena A, Bassetti M, Patroniti N, Schenone A, Pelosi P, Rocco PRM, Robba C. Neurological Manifestations of Severe SARS-CoV-2 Infection: Potential Mechanisms and Implications of Individualized Mechanical Ventilation Settings. Front Neurol 2020; 11:845. [PMID: 32903391 PMCID: PMC7434832 DOI: 10.3389/fneur.2020.00845] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022] Open
Abstract
In December 2019, an outbreak of illness caused by a novel coronavirus (2019-nCoV, subsequently renamed SARS-CoV-2) was reported in Wuhan, China. Coronavirus disease 2019 (COVID-19) quickly spread worldwide to become a pandemic. Typical manifestations of COVID-19 include fever, dry cough, fatigue, and respiratory distress. In addition, both the central and peripheral nervous system can be affected by SARS-CoV-2 infection. These neurological changes may be caused by viral neurotropism, by a hyperinflammatory and hypercoagulative state, or even by mechanical ventilation-associated impairment. Hypoxia, endothelial cell damage, and the different impacts of different ventilatory strategies may all lead to increased stress and strain, potentially exacerbating the inflammatory response and leading to a complex interaction between the lungs and the brain. To date, no studies have taken into consideration the possible secondary effect of mechanical ventilation on brain recovery and outcomes. The aim of our review is to provide an updated overview of the potential pathogenic mechanisms of neurological manifestations in COVID-19, discuss the physiological issues related to brain-lung interactions, and propose strategies for optimization of respiratory support in critically ill patients with SARS-CoV-2 pneumonia.
Collapse
Affiliation(s)
- Denise Battaglini
- Department of Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Iole Brunetti
- Department of Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Pasquale Anania
- Department of Neurosurgery, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Pietro Fiaschi
- Department of Neurosurgery, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Gianluigi Zona
- Department of Neurosurgery, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Lorenzo Ball
- Department of Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Daniele Roberto Giacobbe
- Infectious Disease Unit, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Antonio Vena
- Infectious Disease Unit, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Matteo Bassetti
- Infectious Disease Unit, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Nicolò Patroniti
- Department of Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Angelo Schenone
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Department of Neurology, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Paolo Pelosi
- Department of Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Ministry of Science, Technology, and Innovation, Brasília, Brazil
- Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Chiara Robba
- Department of Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| |
Collapse
|
25
|
Zubair AS, McAlpine LS, Gardin T, Farhadian S, Kuruvilla DE, Spudich S. Neuropathogenesis and Neurologic Manifestations of the Coronaviruses in the Age of Coronavirus Disease 2019: A Review. JAMA Neurol 2020; 77:1018-1027. [PMID: 32469387 PMCID: PMC7484225 DOI: 10.1001/jamaneurol.2020.2065] [Citation(s) in RCA: 642] [Impact Index Per Article: 128.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 2019, causing human coronavirus disease 2019 (COVID-19), which has now spread into a worldwide pandemic. The pulmonary manifestations of COVID-19 have been well described in the literature. Two similar human coronaviruses that cause Middle East respiratory syndrome (MERS-CoV) and severe acute respiratory syndrome (SARS-CoV-1) are known to cause disease in the central and peripheral nervous systems. Emerging evidence suggests COVID-19 has neurologic consequences as well. Observations This review serves to summarize available information regarding coronaviruses in the nervous system, identify the potential tissue targets and routes of entry of SARS-CoV-2 into the central nervous system, and describe the range of clinical neurological complications that have been reported thus far in COVID-19 and their potential pathogenesis. Viral neuroinvasion may be achieved by several routes, including transsynaptic transfer across infected neurons, entry via the olfactory nerve, infection of vascular endothelium, or leukocyte migration across the blood-brain barrier. The most common neurologic complaints in COVID-19 are anosmia, ageusia, and headache, but other diseases, such as stroke, impairment of consciousness, seizure, and encephalopathy, have also been reported. Conclusions and Relevance Recognition and understanding of the range of neurological disorders associated with COVID-19 may lead to improved clinical outcomes and better treatment algorithms. Further neuropathological studies will be crucial to understanding the pathogenesis of the disease in the central nervous system, and longitudinal neurologic and cognitive assessment of individuals after recovery from COVID-19 will be crucial to understand the natural history of COVID-19 in the central nervous system and monitor for any long-term neurologic sequelae.
Collapse
Affiliation(s)
- Adeel S Zubair
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Lindsay S McAlpine
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Tova Gardin
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Shelli Farhadian
- Division of Infectious Disease, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
- Division of Neurological Infections and Global Neurology, Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Deena E Kuruvilla
- Division of Headache and Facial Pain, Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Serena Spudich
- Division of Neurological Infections and Global Neurology, Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
26
|
SARS-CoV-2 and the Nervous System: From Clinical Features to Molecular Mechanisms. Int J Mol Sci 2020. [PMID: 32751841 DOI: 10.3390/ijms21155475.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Increasing evidence suggests that Severe Acute Respiratory Syndrome-coronavirus-2 (SARS-CoV-2) can also invade the central nervous system (CNS). However, findings available on its neurological manifestations and their pathogenic mechanisms have not yet been systematically addressed. A literature search on neurological complications reported in patients with COVID-19 until June 2020 produced a total of 23 studies. Overall, these papers report that patients may exhibit a wide range of neurological manifestations, including encephalopathy, encephalitis, seizures, cerebrovascular events, acute polyneuropathy, headache, hypogeusia, and hyposmia, as well as some non-specific symptoms. Whether these features can be an indirect and unspecific consequence of the pulmonary disease or a generalized inflammatory state on the CNS remains to be determined; also, they may rather reflect direct SARS-CoV-2-related neuronal damage. Hematogenous versus transsynaptic propagation, the role of the angiotensin II converting enzyme receptor-2, the spread across the blood-brain barrier, the impact of the hyperimmune response (the so-called "cytokine storm"), and the possibility of virus persistence within some CNS resident cells are still debated. The different levels and severity of neurotropism and neurovirulence in patients with COVID-19 might be explained by a combination of viral and host factors and by their interaction.
Collapse
|
27
|
Pennisi M, Lanza G, Falzone L, Fisicaro F, Ferri R, Bella R. SARS-CoV-2 and the Nervous System: From Clinical Features to Molecular Mechanisms. Int J Mol Sci 2020; 21:E5475. [PMID: 32751841 PMCID: PMC7432482 DOI: 10.3390/ijms21155475] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Increasing evidence suggests that Severe Acute Respiratory Syndrome-coronavirus-2 (SARS-CoV-2) can also invade the central nervous system (CNS). However, findings available on its neurological manifestations and their pathogenic mechanisms have not yet been systematically addressed. A literature search on neurological complications reported in patients with COVID-19 until June 2020 produced a total of 23 studies. Overall, these papers report that patients may exhibit a wide range of neurological manifestations, including encephalopathy, encephalitis, seizures, cerebrovascular events, acute polyneuropathy, headache, hypogeusia, and hyposmia, as well as some non-specific symptoms. Whether these features can be an indirect and unspecific consequence of the pulmonary disease or a generalized inflammatory state on the CNS remains to be determined; also, they may rather reflect direct SARS-CoV-2-related neuronal damage. Hematogenous versus transsynaptic propagation, the role of the angiotensin II converting enzyme receptor-2, the spread across the blood-brain barrier, the impact of the hyperimmune response (the so-called "cytokine storm"), and the possibility of virus persistence within some CNS resident cells are still debated. The different levels and severity of neurotropism and neurovirulence in patients with COVID-19 might be explained by a combination of viral and host factors and by their interaction.
Collapse
Affiliation(s)
- Manuela Pennisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia, 97–95123 Catania, Italy; (M.P.); (F.F.)
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Via Santa Sofia, 78–95123 Catania, Italy
- Oasi Research Institute–IRCCS, Via Conte Ruggero, 73–94018 Troina, Italy;
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori “Fondazione G. Pascale”, Via Mariano Semmola, 53 –80131 Naples, Italy;
| | - Francesco Fisicaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia, 97–95123 Catania, Italy; (M.P.); (F.F.)
| | - Raffaele Ferri
- Oasi Research Institute–IRCCS, Via Conte Ruggero, 73–94018 Troina, Italy;
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, Via Santa Sofia, 87–95123 Catania, Italy;
| |
Collapse
|
28
|
Abstract
Cardiovascular disease and infections are major causes for the high incidence of morbidity and mortality of patients with chronic kidney disease. Both complications are directly or indirectly associated with disturbed functions or altered apoptotic rates of polymorphonuclear leukocytes, monocytes, lymphocytes, and dendritic cells. Normal responses of immune cells can be reduced, leading to infectious diseases or pre-activated/primed, giving rise to inflammation and subsequently to cardiovascular disease. This review summarizes the impact of kidney dysfunction on the immune system. Renal failure results in disturbed renal metabolic activities with reduced renin, erythropoietin, and vitamin D production, which adversely affects the immune system. Decreased kidney function also leads to reduced glomerular filtration and the retention of uremic toxins. A large number of uremic toxins with detrimental effects on immune cells have been identified. Besides small water-soluble and protein-bound compounds originating from the intestinal microbiome, several molecules in the middle molecular range, e.g., immunoglobulin light chains, retinol-binding protein, the neuropeptides Met-enkephalin and neuropeptide Y, endothelin-1, and the adipokines leptin and resistin, adversely affect immune cells. Posttranslational modifications such as carbamoylation, advanced glycation products, and oxidative modifications contribute to uremic toxicity. Furthermore, high-density lipoprotein from uremic patients has an altered protein profile and thereby loses its anti-inflammatory properties.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
29
|
Girndt M, Trojanowicz B, Ulrich C. Monocytes in Uremia. Toxins (Basel) 2020; 12:toxins12050340. [PMID: 32455723 PMCID: PMC7290468 DOI: 10.3390/toxins12050340] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Monocytes play an important role in both innate immunity and antigen presentation for specific cellular immune defense. In patients with chronic renal failure, as well as those treated with maintenance hemodialysis, these cells are largely dysregulated. There is a large body of literature on monocyte alterations in such patients. However, most of the publications report on small series, there is a vast spectrum of different methods and the heterogeneity of the data prevents any meta-analytic approach. Thus, a narrative review was performed to describe the current knowledge. Monocytes from patients with chronic renal failure differ from those of healthy individuals in the pattern of surface molecule expression, cytokine and mediator production, and function. If these findings can be summarized at all, they might be subsumed as showing chronic inflammation in resting cells together with limited activation upon immunologic challenge. The picture is complicated by the fact that monocytes fall into morphologically and functionally different populations and population shifts interact heavily with dysregulation of the individual cells. Severe complications of chronic renal failure such as impaired immune defense, inflammation, and atherosclerosis can be related to several aspects of monocyte dysfunction. Therefore, this review aims to provide an overview about the impairment and activation of monocytes by uremia and the resulting clinical consequences for renal failure patients.
Collapse
|
30
|
Musso G, De Michieli F, Bongiovanni D, Parente R, Framarin L, Leone N, Berrutti M, Gambino R, Cassader M, Cohney S, Paschetta E. New Pharmacologic Agents That Target Inflammation and Fibrosis in Nonalcoholic Steatohepatitis-Related Kidney Disease. Clin Gastroenterol Hepatol 2017; 15:972-985. [PMID: 27521506 DOI: 10.1016/j.cgh.2016.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023]
Abstract
Epidemiologic data show an association between the prevalence and severity of nonalcoholic fatty liver disease and the incidence and stage of chronic kidney disease (CKD); furthermore, nonalcoholic steatohepatitis (NASH)-related cirrhosis has a higher risk of renal failure, a greater necessity for simultaneous liver-kidney transplantation, and a poorer renal outcome than cirrhosis of other etiologies even after simultaneous liver-kidney transplantation. These data suggest that NASH and CKD share common proinflammatory and profibrotic mechanisms of progression, which are targeted incompletely by current treatments. We reviewed therapeutic approaches to late preclinical/early clinical stage of development in NASH and/or CKD, focusing on anti-inflammatory and antifibrotic treatments, which could slow the progression of both disease conditions. Renin inhibitors and angiotensin-converting enzyme-2 activators are new renin-angiotensin axis modulators that showed incremental advantages over angiotensin-converting enzyme inhibitors/angiotensin-receptor blockers in preclinical models. Novel, potent, and selective agonists of peroxisome proliferator-activated receptors and of farnesoid X receptor, designed to overcome limitations of older compounds, showed promising results in clinical trials. Epigenetics, heat stress response, and common effectors of redox regulation also were subjected to intensive research, and the gut was targeted by several approaches, including synbiotics, antilipopolysaccharide antibodies, Toll-like receptor-4 antagonists, incretin mimetics, and fibroblast growth factor 19 analogs. Promising anti-inflammatory therapies include inhibitors of NOD-like receptor family, pyrin domain containing 3 inflammasome, of nuclear factor-κB, and of vascular adhesion protein-1, chemokine antagonists, and solithromycin, and approaches targeting common profibrogenic pathways operating in the liver and the kidney include galectin-3 antagonists, and inhibitors of rho-associated protein kinase and of epidermal growth factor activation. The evidence, merits, and limitations of each approach for the treatment of NASH and CKD are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicola Leone
- Gradenigo Hospital, University of Turin, Turin, Italy
| | - Mara Berrutti
- Gradenigo Hospital, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Maurizio Cassader
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Solomon Cohney
- Department of Nephrology, Royal Melbourne and Western Hospital, Victoria, University of Melbourne, Australia
| | | |
Collapse
|
31
|
Kim HY, Yoo TH, Hwang Y, Lee GH, Kim B, Jang J, Yu HT, Kim MC, Cho JY, Lee CJ, Kim HC, Park S, Lee WW. Indoxyl sulfate (IS)-mediated immune dysfunction provokes endothelial damage in patients with end-stage renal disease (ESRD). Sci Rep 2017; 7:3057. [PMID: 28596556 PMCID: PMC5465082 DOI: 10.1038/s41598-017-03130-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
Progressive renal failure causes uremia-related immune dysfunction, which features a chronic inflammatory milieu. Given the central role of end-stage renal disease (ESRD)-related immune dysfunction in the pathogenesis of cardiovascular diseases (CVDs), much attention has been focused on how uremic toxins affect cellular immunity and the mechanisms underlying pathogenesis of atherosclerosis in ESRD patients. Here, we investigated the characteristics of monocytes and CD4+ T cells in ESRD patients and the immune responses induced by indoxyl sulfate (IS), a key uremic toxin, in order to explore the pathogenic effects of these cells on vascular endothelial cells. In ESRD patients, monocytes respond to IS through the aryl hydrocarbon receptor (AhR) and consequently produce increased levels of TNF-α. Upon stimulation with TNF-α, human vascular endothelial cells produce copious amounts of CX3CL1, a chemokine ligand of CX3CR1 that is highly expressed on CD4+CD28-T cells, the predominantly expanded cell type in ESRD patients. A migration assay showed that CD4+CD28- T cells were preferentially recruited by CX3CL1. Moreover, activated CD4+CD28- T cells exhibited cytotoxic capability allowing for the induction of apoptosis in HUVECs. Our findings suggest that in ESRD, IS-mediated immune dysfunction may cause vascular endothelial cell damage and thus, this toxin plays a pivotal role in the pathogenesis of CVD.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae-Hyun Yoo
- Division of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yuri Hwang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Ga Hye Lee
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Bonah Kim
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Jiyeon Jang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee Tae Yu
- Division of Cardiology, Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Chang Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Chan Joo Lee
- Department of Health Promotion and Disease Prevention, Severance Hospital, Seoul, South Korea
| | - Hyeon Chang Kim
- Cardiovascular and Metabolic Diseases Etiology Research Center and Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sungha Park
- Division of Cardiology, Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea.
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Ischemic/Hypoxic Disease Institute and Institute of Infectious Diseases, Seoul National University College of Medicine; Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea.
| |
Collapse
|
32
|
Pošćić N, Montanari T, D’Andrea M, Licastro D, Pilla F, Ajmone-Marsan P, Minuti A, Sgorlon S. Breed and adaptive response modulate bovine peripheral blood cells' transcriptome. J Anim Sci Biotechnol 2017; 8:11. [PMID: 28149510 PMCID: PMC5264304 DOI: 10.1186/s40104-017-0143-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/07/2017] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Adaptive response includes a variety of physiological modifications to face changes in external or internal conditions and adapt to a new situation. The acute phase proteins (APPs) are reactants synthesized against environmental stimuli like stress, infection, inflammation. METHODS To delineate the differences in molecular constituents of adaptive response to the environment we performed the whole-blood transcriptome analysis in Italian Holstein (IH) and Italian Simmental (IS) breeds. For this, 663 IH and IS cows from six commercial farms were clustered according to the blood level of APPs. Ten extreme individuals (five APP+ and APP- variants) from each farm were selected for the RNA-seq using the Illumina sequencing technology. Differentially expressed (DE) genes were analyzed using dynamic impact approach (DIA) and DAVID annotation clustering. Milk production data were statistically elaborated to assess the association of APP+ and APP- gene expression patterns with variations in milk parameters. RESULTS The overall de novo assembly of cDNA sequence data generated 13,665 genes expressed in bovine blood cells. Comparative genomic analysis revealed 1,152 DE genes in the comparison of all APP+ vs. all APP- variants; 531 and 217 DE genes specific for IH and IS comparison respectively. In all comparisons overexpressed genes were more represented than underexpressed ones. DAVID analysis revealed 369 DE genes across breeds, 173 and 73 DE genes in IH and IS comparison respectively. Among the most impacted pathways for both breeds were vitamin B6 metabolism, folate biosynthesis, nitrogen metabolism and linoleic acid metabolism. CONCLUSIONS Both DIA and DAVID approaches produced a high number of significantly impacted genes and pathways with a narrow connection to adaptive response in cows with high level of blood APPs. A similar variation in gene expression and impacted pathways between APP+ and APP- variants was found between two studied breeds. Such similarity was also confirmed by annotation clustering of the DE genes. However, IH breed showed higher and more differentiated impacts compared to IS breed and such particular features in the IH adaptive response could be explained by its higher metabolic activity. Variations of milk production data were significantly associated with APP+ and APP- gene expression patterns.
Collapse
Affiliation(s)
- Nataliya Pošćić
- Department of Agriculture, Food, Environment and Animal Science (DI4A), University of Udine, via delle Scienze 206, 33100 Udine, Italy
| | - Tommaso Montanari
- Department of Agriculture, Food, Environment and Animal Science (DI4A), University of Udine, via delle Scienze 206, 33100 Udine, Italy
| | - Mariasilvia D’Andrea
- Department of Agricultural, Environmental and Food Sciences, University of Molise, via F. De Sanctis snc, 86100 Campobasso, Italy
| | - Danilo Licastro
- CBM S.c.r.l, SS 14 – km 163.5 AREA Science Park, 34149 Basovizza, TS Italy
| | - Fabio Pilla
- Department of Agricultural, Environmental and Food Sciences, University of Molise, via F. De Sanctis snc, 86100 Campobasso, Italy
| | - Paolo Ajmone-Marsan
- Institute of Zootechnics, Catholic University of the Sacred Heart, via Emilia Parmense 84, 29133 Piacenza, Italy
| | - Andrea Minuti
- Institute of Zootechnics, Catholic University of the Sacred Heart, via Emilia Parmense 84, 29133 Piacenza, Italy
| | - Sandy Sgorlon
- Department of Agriculture, Food, Environment and Animal Science (DI4A), University of Udine, via delle Scienze 206, 33100 Udine, Italy
| |
Collapse
|
33
|
Úri K, Fagyas M, Kertész A, Borbély A, Jenei C, Bene O, Csanádi Z, Paulus WJ, Édes I, Papp Z, Tóth A, Lizanecz E. Circulating ACE2 activity correlates with cardiovascular disease development. J Renin Angiotensin Aldosterone Syst 2016; 17:17/4/1470320316668435. [PMID: 27965422 PMCID: PMC5843890 DOI: 10.1177/1470320316668435] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/05/2016] [Indexed: 12/17/2022] Open
Abstract
It was shown recently that angiotensin-converting enzyme activity is limited by endogenous inhibition in vivo, highlighting the importance of angiotensin II (ACE2) elimination. The potential contribution of the ACE2 to cardiovascular disease progression was addressed. Serum ACE2 activities were measured in different clinical states (healthy, n=45; hypertensive, n=239; heart failure (HF) with reduced ejection fraction (HFrEF) n=141 and HF with preserved ejection fraction (HFpEF) n=47). ACE2 activity was significantly higher in hypertensive patients (24.8±0.8 U/ml) than that in healthy volunteers (16.2±0.8 U/ml, p=0.01). ACE2 activity further increased in HFrEF patients (43.9±2.1 U/ml, p=0.001) but not in HFpEF patients (24.6±1.9 U/ml) when compared with hypertensive patients. Serum ACE2 activity negatively correlated with left ventricular systolic function in HFrEF, but not in hypertensive, HFpEF or healthy populations. Serum ACE2 activity had a fair diagnostic value to differentiate HFpEF from HFrEF patients in this study. Serum ACE2 activity correlates with cardiovascular disease development: it increases when hypertension develops and further increases when the cardiovascular disease further progresses to systolic dysfunction, suggesting that ACE2 metabolism plays a role in these processes. In contrast, serum ACE2 activity does not change when hypertension progresses to HFpEF, suggesting a different pathomechanism for HFpEF, and proposing a biomarker-based identification of these HF forms.
Collapse
Affiliation(s)
- Katalin Úri
- Department of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Fagyas
- Department of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Kertész
- Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Borbély
- Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Jenei
- Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Orsolya Bene
- Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Csanádi
- Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Walter J Paulus
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, the Netherlands
| | - István Édes
- Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Department of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Tóth
- Department of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Erzsébet Lizanecz
- Department of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary .,Department of Cardiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|