1
|
Azcarate D, Olasagasti Arsuaga F, Granizo Rodriguez E, Arana-Arri E, España PP, Intxausti M, Sancho C, García de Vicuña Meléndez A, Ibarrondo O, M de Pancorbo M. Human-genetic variants associated with susceptibility to SARS-CoV-2 infection. Gene 2025; 953:149423. [PMID: 40120867 DOI: 10.1016/j.gene.2025.149423] [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: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
SARS-CoV-2, the third major coronavirus of the 21st century, causing COVID-19 disease, profoundly impacts public health and workforces worldwide. Identifying individuals at heightened risk of SARS-CoV-2 infection is crucial for targeted interventions and preparedness. This study investigated 35 SNVs within viral infection-associated genes in SARS-CoV-2 patients and uninfected controls from the Basque Country (March 2020-July 2021). Its primary aim was to uncover genetic markers indicative of SARS-CoV-2 susceptibility and explore genetic predispositions to infection. Association analyses revealed previously unreported associations between SNVs and susceptibility. Haplotype analyses uncovered novel links between haplotypes and susceptibility, surpassing individual SNV associations. Descriptive modelling identified key susceptibility factors, with rs11246068-CC (IFITM3), rs5742933-GG (ORMDL1), rs35337543-CG (IFIH1), and GGGCT (rs2070788, rs2298659, rs17854725, rs12329760, rs3787950) variation in TMPRSS2 emerging as main infection-susceptibility indicators for a COVID-19 pandemic situation. These findings underscore the importance of integrated SNV and haplotype analyses in delineating susceptibility to SARS-CoV-2 and informing proactive prevention strategies. The genetic markers profiled in this study offer valuable insights for future pandemic preparedness.
Collapse
Affiliation(s)
- Daniel Azcarate
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Felix Olasagasti Arsuaga
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Biochemistry and molecular biology department, Faculty of Pharmacy (UPV/EHU), 01006 Vitoria-Gasteiz, Alava (Basque Country), Spain.
| | - Eva Granizo Rodriguez
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Eunate Arana-Arri
- Clinical Epidemiology Unit, Cruces University Hospital, 48903 Barakaldo, Biscay (Basque Country), Spain
| | - Pedro Pablo España
- Pulmonology Service, Galdakao-Usansolo University Hospital, 48960 Galdakao, Biscay (Basque Country), Spain
| | - Maider Intxausti
- Pulmonology Service, Alava University Hospital - Txagorritxu, 01009 Vitoria-Gasteiz, Álava (Basque Country), Spain
| | - Cristina Sancho
- Department of Pneumology, Basurto University Hospital, 48013 Bilbao, Biscay (Basque Country), Spain
| | | | - Oliver Ibarrondo
- Consultant in Statistics and Health Economics Research, Debagoiena AP-OSI Research Unit, 20500 Arrasate, Gipuzkoa (Basque Country), Spain
| | - Marian M de Pancorbo
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain.
| |
Collapse
|
2
|
Yang L, Zhou X, Liu J, Yang G, Tan W, Ding H, Fang X, Yu J, Li W, He J, Cao H, Ma Q, Yu L, Lu Z. PEBL, a component-based Chinese medicine, reduces virus-induced acute lung injury by targeting FXR to decrease ACE2 levels. J Adv Res 2025:S2090-1232(25)00295-4. [PMID: 40324631 DOI: 10.1016/j.jare.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/16/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION Despite the growing clinical need, the therapeutic efficacy of drugs for acute lung injury (ALI) remains inadequate. Traditional Chinese Medicine (TCM) holds potential in managing ALI due to its unique therapeutic properties. However, the intricate nature of TCM formulations hinders global adoption. Component-based Chinese medicine (CCM) offers a promising pathway for TCM's internationalization. Phillyrin-Emodin-Baicalin-Liquiritin (PEBL), a CCM with significant anti-inflammatory activity, is derived from the well-established TCM formula Liang-Ge-San. Whether PEBL effectively addresses viral ALI, however, remains unclear. OBJECTIVES This study aims to investigate the therapeutic effects and underlying mechanisms of PEBL on viral ALI. METHODS The efficacy of PEBL against Poly(I:C)-induced ALI was assessed by analyzing cytokine production, macrophage infiltration, pulmonary damage, and mortality. Bioinformatics and network pharmacology were employed to identify key targets and signaling pathways. The molecular mechanisms were further validated using Poly(I:C)-treated RAW264.7 cells, Tg(coro1α: GFP) zebrafish, BALB/c mice, and models of Influenza A/Puerto Rico/8/1934 (H1N1) virus strain (PR8)-induced ALI in BALB/c mice and SARS-CoV-2 Omicron XBB.1.16 subvariant (XBB)-induced ALI in hACE2-transgenic C57BL/6 mice. RESULTS PEBL mitigated Poly(I:C)-induced ALI, as evidenced by reduced cytokine levels, diminished macrophage infiltration, alleviated lung damage, and decreased mortality. Virtual screening identified the farnesyl X receptor (FXR) and angiotensin-converting enzyme 2 (ACE2) as key therapeutic targets for viral pneumonia. Mechanistically, PEBL downregulated FXR expression, inhibiting FXR binding to ACE2 promoters, which subsequently suppressed NF-κB-p65 nuclear translocation and cytokine production. In vivo, PEBL attenuated cytokine production by inhibiting ACE2 transcription through FXR downregulation, leading to alleviation of Poly(I:C)-induced ALI in both zebrafish and mice. Additionally, PEBL significantly improved symptoms of ALI caused by PR8 and XBB infections, by disrupting the FXR/ACE2 signaling axis, resulting in reduced weight loss, lower lung indices, diminished viral load and titer, fewer pulmonary lesions, and suppressed NF-κB-p65 nuclear translocation, along with decreased cytokine storm. CONCLUSIONS This study provides the first evidence that PEBL offers protective effects against ALI induced by acute respiratory viruses. PEBL prevents FXR from binding to ACE2 by inhibiting FXR transcription, which reduces macrophage infiltration, cytokine storm formation, and inflammatory injury, thereby ameliorating viral ALI. These findings underscore the potential of PEBL as a candidate for further exploration in the treatment of viral ALI.
Collapse
Affiliation(s)
- Liling Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Guangli Yang
- Department of Central Laboratory, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Weifu Tan
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Hongyan Ding
- Omega-3 Research and Conversion Center, Dongguan Innovation Research Institute, Guangdong Medical University, Dongguan 523808, China
| | - Xiaochuan Fang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wei Li
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Jiayang He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, China
| | - Huihui Cao
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
3
|
Vanderkamp SG, Niazy M, Stegelmeier AA, Stinson KJ, Ricker N, Bridle BW. Cytokine, chemokine, and acute-phase protein profiles in plasma as correlative biomarkers of clinical outcomes for patients with COVID-19. Sci Rep 2025; 15:15397. [PMID: 40316702 PMCID: PMC12048561 DOI: 10.1038/s41598-025-99248-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025] Open
Abstract
Coronavirus disease identified in 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2, had a global impact on human health and the economy. The aim of this study was to quantify cytokines, chemokines, and acute phase proteins in the plasma of patients with COVID-19 to elucidate potential biomarkers to inform prognostic and treatment decisions. Clustering analysis using the K-prototypes method identified underlying biological patterns in patients with COVID-19. The penalized multinomial logistic regression analysis identified two comorbidities (hypertension, congestive heart failure) and thirteen analytes as potential risk factors for COVID-19 progression with 88.2% accuracy. Based on a patient's age, high concentrations of interleukin (IL)-6, monocyte chemoattractant protein-1, and pentraxin 3 were important biomarkers for lethal COVID-19. Decreased concentrations of interferon gamma-induced protein-10, IL-10, and soluble tumor necrosis factor receptor I were found to be associated with mild COVID-19, while increasing concentrations of these analytes could be used to predict COVID-19 severity.
Collapse
Affiliation(s)
- Sierra G Vanderkamp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Maysa Niazy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Nicole Ricker
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
4
|
Icoz SGG, Yorgun MA, Bayhan GI, Icoz M, Yahsi A. Ocular hemodynamics in multisystem inflammatory syndrome in children: A cross-sectional study. Indian J Ophthalmol 2025; 73:725-730. [PMID: 39728610 DOI: 10.4103/ijo.ijo_1527_24] [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: 06/27/2024] [Accepted: 10/27/2024] [Indexed: 12/28/2024] Open
Abstract
PURPOSE To evaluate retinal vascular changes by optical coherence tomography angiography (OCTA) in multisystem inflammatory syndrome in children (MIS-C). METHODS This cross-sectional study included 21 patients who were diagnosed with MIS-C and had a history of hospitalization, 20 pediatric outpatients with a coronavirus disease 2019 (COVID-19) diagnosis, and 26 healthy children. All patients underwent a detailed ophthalmologic examination and OCTA. In the MIS-C and pediatric COVID-19 groups, these evaluations were made 6 months after diagnosis. The vascular density values of the superficial, deep, and radial peripapillary capillary plexuses (SCP, DCP, and RPCP, respectively), foveal avascular zone (FAZ) parameters (area, perimeter, acircularity index, and foveal density), and outer retinal and choriocapillaris flow area values were recorded using OCTA. RESULTS No pathology was detected in the ophthalmologic examinations of the three groups with similar age and gender distributions. Although the vascular density values of SCP, DCP, and RPCP were found to be higher in most quadrants in the MIS-C group, there was no statistically significant difference among the three groups ( P > 0.05 for all). FAZ parameters and flow area measurements were similar in all three groups ( P > 0.05 for all). CONCLUSION This is the first study to evaluate relatively long-term outcomes in patients with MIS-C and pediatric COVID-19 together. This study shows no changes in the SCP and DCP parameters in pediatric age group, which shows that ocular hemodynamic changes may not be reflected on OCTA after 6 months.
Collapse
Affiliation(s)
| | - Mucella Arıkan Yorgun
- Department of Ophthalmology, Yıldırım Beyazıt University Faculty of Medicine, Ankara, Türkiye
| | - Gulsum Iclal Bayhan
- Division of Pediatric Infectious Disease, Ankara City Hospital, Ankara Yıldırım Beyazıt University Faculty of Medicine, Ankara, Türkiye
| | - Mehmet Icoz
- Department of Ophthalmology, Yozgat City Hospital, Yozgat, Türkiye
| | - Aysun Yahsi
- Division of Pediatric Infectious Disease, Ankara City Hospital, Ankara Yıldırım Beyazıt University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
5
|
Hachimi A, El-Mansoury B, Merzouki M. Incidence, pathophysiology, risk factors, histopathology, and outcomes of COVID-19-induced acute kidney injury: A narrative review. Microb Pathog 2025; 202:107360. [PMID: 39894232 DOI: 10.1016/j.micpath.2025.107360] [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: 09/03/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has led to a significant burden on global healthcare systems. COVID-19-induced acute kidney injury (AKI) is among one of the complications, that has emerged as a critical and frequent condition in COVID-19 patients. This AKI among COVID-19 patients is associated with poor outcomes, and high mortality rates, especially in those with severe AKI or requiring renal replacement therapy. COVID-19-induced AKI represents a significant complication with complex pathophysiology and multifactorial risk factors. Indeed, several pathophysiological mechanisms, including direct viral invasion of renal cells, systemic inflammation, endothelial and thrombotic abnormalities as well as nephrotoxic drugs and rhabdomyolysis are believed to underlie this condition. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include acute tubular necrosis, glomerular injury, and the presence of viral particles within renal tissue and urine. Identified risk factors for developing AKI vary among studies, depending on regions, underlying conditions, and the severity of the disease. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include show acute tubular necrosis, glomerular injury, and viral particles within renal tissue and urine. While, identified risk factors for developing AKI vary among studies, according to regions, underlying conditions, and the gravity of the disease. This narrative review aims to synthesize current knowledge on the incidence, pathophysiology, risk factors, histopathology, and outcomes of AKI induced by COVID-19.
Collapse
Affiliation(s)
- Abdelhamid Hachimi
- Medical ICU, Mohammed VI(th) University Hospital of Marrakech, Marrakech, Morocco; Morpho-Science Research Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco; Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Bilal El-Mansoury
- Nutritional Physiopathologies, Neuroscience and Toxicology Team, Laboratory of Anthropogenic, Biotechnology and Health, Faculty of Sciences, Chouaib Doukkali University, El Jadida, Morocco
| | - Mohamed Merzouki
- Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco.
| |
Collapse
|
6
|
Dong D, Song Y, Wu S, Wang B, Peng C, Zhang W, Kong W, Zhang Z, Song J, Hou LH, Li S. Molecular basis of Ad5-nCoV vaccine-induced immunogenicity. Structure 2025; 33:858-868.e5. [PMID: 40112804 DOI: 10.1016/j.str.2025.02.009] [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: 07/18/2024] [Revised: 12/12/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Ad5-nCoV (Convidecia) is listed for emergency use against COVID-19 by the World Health Organization (WHO) and has been globally administered to millions of people. It utilizes human adenovirus 5 (Ad5) replication-incompetent vector to deliver the spike (S) protein gene from various SARS-CoV-2 strains. Despite promising clinical data, the molecular mechanism underlying its high immunogenicity and adverse reactions remain incompletely understood. Here, we primarily applied cryo-electron tomography (cryo-ET), fluorescence microscopy and mass spectrometry to analyze the Ad5-nCoV_Wu and Ad5-nCoV_O vaccine-induced S antigens. These antigens encode the unmodified SARS-CoV-2 Wuhan-Hu-1 S gene and the stabilized Omicron S gene, respectively. Our findings highlight the structural integrity, antigenicity, and dense distribution on cell membrane of the vaccine-induced S proteins. Ad5-nCoV_O induced S proteins exhibit improved stability and reduced syncytia formation among inoculated cells. Our work demonstrates that Ad5-nCoV is a prominent platform for antigen induction and cryo-ET can be a useful technique for vaccine characterization and development.
Collapse
Affiliation(s)
- Dongyang Dong
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yutong Song
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shipo Wu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Busen Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Cheng Peng
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weiping Zhang
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weizheng Kong
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zheyuan Zhang
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jingwen Song
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Li-Hua Hou
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Sai Li
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
7
|
Mansilla-Sandoval A, Corrales-Delgado D, Puyén ZM, Mansilla-Doria P, Orendo-Velásquez E, Huicho L, Fano-Sizgorich D. SARS-CoV-2 infection and complicated appendicitis in adults in Lima, Peru: a matched case-control study. BMC Surg 2025; 25:159. [PMID: 40234792 PMCID: PMC12001637 DOI: 10.1186/s12893-025-02897-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Acute appendicitis may be uncomplicated or may present with life threatening complications. Since the outbreak of the COVID-19 pandemic, there has been an increase in the number of cases of complicated appendicitis, suggesting a possible association between them. Therefore, we aimed to determine the association between SARS-CoV-2 infection and complicated appendicitis in surgical patients in Lima, Peru, from March 2020 to December 2021. METHODS A matched case-control study was conducted. Clinical records of patients ≥ 18 years old who underwent surgery for appendicitis and had at least one positive SARS-CoV-2 diagnostic test were selected. Patients undergoing surgery for complicated appendicitis were considered cases, and patients undergoing surgery for uncomplicated appendicitis were controls. A 1:1 matching by sex, age, and month of surgery was performed. Conditional logistic regression modeling was performed to calculate crude and adjusted conditional odds ratios (cOR). RESULTS The positivity rate for COVID-19 tests was 73.6% for cases and 26.4% for controls. The crude cOR was 4.88 (95% IC 2.89-8.23, p < 0.001), and the adjusted cOR was 3.52 (95%IC 1.82-6.81, p = 0.001), after controlling for onset time of symptoms and awaiting time before surgery. CONCLUSIONS Surgery for complicated appendicitis was associated with SARS-CoV-2 infection. Patients with this infection may be at higher risk of complicated appendicitis and thus may need additional clinical monitoring.
Collapse
Affiliation(s)
| | | | - Zully M Puyén
- Facultad de Ciencias de la Salud, Universidad Peruana de Ciencias Aplicadas, Lima, Peru
| | - Percy Mansilla-Doria
- Facultad de Ciencias de la Salud, Universidad Peruana de Ciencias Aplicadas, Lima, Peru
- Servicio de Cirugía General, Hospital de Emergencias Grau- EsSalud, Lima, Peru
| | | | - Luis Huicho
- Centro de Investigación en Salud Materna e Infantil, Centro de Investigación para el Desarrollo Integral y Sostenible and Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Diego Fano-Sizgorich
- Laboratorio de Endocrinología y Reproducción, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru.
| |
Collapse
|
8
|
Mitsutake A, Sakai Y, Tsuboyama Y, Baba Y, Arakawa A, Saito Y, Tsumoto M, Ikeda S, Iwata NK. Bilateral Middle Cerebellar Peduncle Sign in a Patient with Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy and Coronavirus Disease 2019. Intern Med 2025; 64:1263-1266. [PMID: 39924233 DOI: 10.2169/internalmedicine.4826-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2025] Open
Abstract
A 59-year-old man without risk factors for atherosclerosis was diagnosed with coronavirus disease 2019 (COVID-19). Four days later, he developed dysarthria and gait disturbance. Neurological examination revealed slurred speech, ataxia, and mild cognitive decline. Brain magnetic resonance imaging revealed multiple infarcts in the bilateral middle cerebellar peduncles and leukoencephalopathy, indicating the diagnosis of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Genetic testing confirmed a pathogenic NOTCH3 variant (c.505C>T, p.Arg169Cys) (NM_000435.3). A skin biopsy supported the diagnosis. He was treated with cilostazol and after three months of rehabilitation, he regained an independent walking ability. COVID-19 increases the risk of ischemic stroke in CADASIL patients, with bilateral middle cerebellar peduncle infarctions being notable in the present case.
Collapse
Affiliation(s)
- Akihiko Mitsutake
- Department of Neurology, International University of Health and Welfare Mita Hospital, Japan
| | - Yuto Sakai
- Department of Neurology, International University of Health and Welfare Mita Hospital, Japan
| | - Yoko Tsuboyama
- Department of Neurology, International University of Health and Welfare Mita Hospital, Japan
| | - Yusuke Baba
- Department of Neurology, International University of Health and Welfare Mita Hospital, Japan
| | - Akira Arakawa
- Department of Neurology and Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan
| | - Yuko Saito
- Department of Neurology and Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan
| | - Manabu Tsumoto
- Department of Neurosurgery, JCHO Tokyo Takanawa Hospital, Japan
| | - Shigeru Ikeda
- Department of Neurology, JCHO Tokyo Takanawa Hospital, Japan
| | - Nobue K Iwata
- Department of Neurology, International University of Health and Welfare Mita Hospital, Japan
| |
Collapse
|
9
|
Gintoni I, Mastrogeorgiou M, Papakosta V, Vassiliou S, Yapijakis C. Genetic Variations Related to Angiotensin II Production and Risk for Basal Cell Carcinoma. Biochem Genet 2025; 63:917-935. [PMID: 38546913 DOI: 10.1007/s10528-024-10746-0] [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: 05/30/2023] [Accepted: 02/16/2024] [Indexed: 03/23/2025]
Abstract
Basal cell carcinoma (BCC) is the most prevalent human neoplasm, with constantly increasing annual incidence. Despite its slow growth, BCC is locally invasive and, if left untreated, can cause severe complications, including metastasis and death. The renin-angiotensin system (RAS) plays a key role in electrolyte balance, atrial pressure, tissue development, homeostasis, and inflammation, but also in cancer development. After binding to its type 1 receptor (AT1R), angiotensin II (ANGII), the system's principal hormonal effector, regulates cancer pathways spanning from the formation of the initial cancer cell to the construction and nutrition of the tumor microenvironment, angiogenesis, proliferation, and metastasis. Although the role of RAS in the development of skin pathologies has not been widely researched, RAS-targeting antihypertensive medications have been shown to have a chemoprotective effect against BCC. Based on those findings, our group conducted a series of genetic association studies to investigate the association between common functional variations in key genes related to ANGII production (AGT, ACE, ACE2, AT1R, AT2R, and CMA1) and the risk of BCC occurrence. This review provides a summary of the current understanding of the ANGII involvement in BCC development. The reliable and easily assessed pool of genetic biomarkers may be used for predictive testing and prevention purposes in high-risk individuals.
Collapse
Affiliation(s)
- Iphigenia Gintoni
- Unit of Orofacial Genetics, 1st Department of Pediatrics, National Kapodistrian University of Athens, University Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Choremion Laboratory "Hagia Sophia" Children's Hospital, Athens, Greece
- Department of Molecular Genetics, Cephalogenetics Center, Philaretou 88, Kallithea, 17675, Athens, Greece
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Michael Mastrogeorgiou
- Unit of Orofacial Genetics, 1st Department of Pediatrics, National Kapodistrian University of Athens, University Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Choremion Laboratory "Hagia Sophia" Children's Hospital, Athens, Greece
| | - Veronica Papakosta
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Stavros Vassiliou
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Christos Yapijakis
- Unit of Orofacial Genetics, 1st Department of Pediatrics, National Kapodistrian University of Athens, University Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Choremion Laboratory "Hagia Sophia" Children's Hospital, Athens, Greece.
- Department of Molecular Genetics, Cephalogenetics Center, Philaretou 88, Kallithea, 17675, Athens, Greece.
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece.
| |
Collapse
|
10
|
Chatterjee G, Saha AK, Khurshid S, Saha A. A Comprehensive Review of the Antioxidant, Antimicrobial, and Therapeutic Efficacies of Black Cumin ( Nigella sativa L.) Seed Oil and Its Thymoquinone. J Med Food 2025; 28:325-339. [PMID: 39807848 DOI: 10.1089/jmf.2024.k.0149] [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] [Indexed: 01/16/2025] Open
Abstract
Black cumin (Nigella sativa L.) (family Ranunculaceae) is a largely utilized therapeutic herb worldwide. This comprehensive review discusses the pharmacological benefits of black cumin seed oil, focusing on its bioactive component thymoquinone (TQ). The review is structured as follows: First, we examine the antimicrobial properties of black cumin oil, followed by an analysis of its antioxidant capabilities. Finally, we explore its therapeutic potential, particularly in neurodegenerative diseases and COVID-19. Phytochemicals from N. sativa have exhibited potential for developing novel preventive and therapeutic strategies against jaundice, gastrointestinal disorders, skin diseases, anorexia, conjunctivitis, dyspepsia, intrinsic hemorrhage, amenorrhea, paralysis, anorexia, rheumatism, diabetes, hypertension, fever, influenza, eczema, asthma, cough, bronchitis, and headache. The broader spectrum of application for N. sativa and its essential bioactives have certainly enhanced the commercial value of this seed oil. TQ, a major constituent of black cumin seed oil, has numerous beneficial properties. Researchers have extensively studied black cumin seed oil and its major component, TQ. These studies have revealed a wide range of pharmacological properties, including anticancer, immunomodulatory, analgesic, antimicrobial, antidiabetic, and anti-inflammatory effects. Additionally, TQ has shown neuroprotective, spasmolytic, bronchodilatory, hepatoprotective, renoprotective, gastroprotective, and antioxidant activities.
Collapse
Affiliation(s)
- Gourab Chatterjee
- Department of Food Technology, Haldia Institute of Technology, Haldia, India
| | - Asit Kumar Saha
- Department of Chemical Engineering, Haldia Institute of Technology, Haldia, India
| | - Shamama Khurshid
- Department of Food Technology, Haldia Institute of Technology, Haldia, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| |
Collapse
|
11
|
Xie X, Zhang Y, Fang Y, Wu J, Li Q. Molecular Basis of High-Blood-Pressure-Enhanced and High-Fever-Temperature-Weakened Receptor-Binding Domain/Peptidase Domain Binding: A Molecular Dynamics Simulation Study. Int J Mol Sci 2025; 26:3250. [PMID: 40244099 PMCID: PMC11989460 DOI: 10.3390/ijms26073250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
The entry and infection of the Severe Acute Respiratory Syndrome Coronavirus 2 virus (SARS-CoV-2) involve recognition and binding of the receptor-binding domain (RBD) of the virus surface spike protein to the peptidase domain (PD) of the host cellular Angiotensin-Converting Enzyme-2 (ACE2) receptor. ACE2 is also involved in normal blood pressure control. An association between hypertension and COVID-19 severity and fatality is evident, but how hypertension predisposes patients diagnosed with COVID-19 to unfavorable outcomes remains unclear. High temperature early during SARS-CoV-2 infection impairs binding to human cells and retards viral progression. Low body temperature can prelude poor prognosis. In this study, all-atom molecular dynamics simulations were performed to examine the effects of high pressure and temperature on RBD/PD binding. A high blood pressure of 940 mmHg enhanced RBD/PD binding. A high temperature above 315 K significantly weakened RBD/PD binding, while a low temperature of 305 K enhanced binding. The curvature of the PD α1-helix and proximity of the PD β3β4-hairpin tip to the RBM motif affected the compactness of the binding interface and, hence, binding affinity. These findings provide novel insights into the underlying mechanisms by which hypertension predisposes patients to unfavorable outcomes in COVID-19 and how an initial high temperature retards viral progression.
Collapse
Affiliation(s)
| | | | | | - Jianhua Wu
- Institute of Biomechanics, School of Biology and Biological Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Panyu District, Guangzhou 510006, China; (X.X.); (Y.Z.); (Y.F.)
| | - Quhuan Li
- Institute of Biomechanics, School of Biology and Biological Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Panyu District, Guangzhou 510006, China; (X.X.); (Y.Z.); (Y.F.)
| |
Collapse
|
12
|
Noettger S, Zech F, Nchioua R, Pastorio C, Jung C, Jacob T, Stenger S, Kirchhoff F. Role of N-linked glycosylation sites in human ACE2 in SARS-CoV-2 and hCoV-NL63 infection. J Virol 2025:e0220224. [PMID: 40152594 DOI: 10.1128/jvi.02202-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a transmembrane protein known for its physiological role in the renin-angiotensin system that also serves as a receptor for entry of SARS-CoV-1, SARS-CoV-2, and the seasonal human coronavirus NL63 (hCoV-NL63). ACE2 contains seven N-linked glycosylation sites. Molecular simulation and binding analyses suggest that some of them are involved in the interaction with the Spike (S) proteins of hCoVs, but their relevance in S-mediated fusion and viral entry is poorly investigated. To address this, we determined the impact of all seven N-linked glycosylation sites in ACE2 on S-mediated SARS-CoV-2 and hCoV-NL63 infection as well as cell-to-cell fusion. We found that all mutant ACE2 proteins are expressed and localized at the cell surface, albeit ACE2 lacks all glycans at decreased levels. On average, changes in T92I, N322A, and N690A, as well as combined mutation of all N-linked glycosylation sites increased endocytic VSVpp infection mediated by early HU-1 as well as Omicron BA.2, BA.5, and XBB.1.5 SARS-CoV-2 S proteins. In comparison, only the lack of glycan at N322 in ACE2 enhanced syncytia formation and only in the case of HU-1 and XBB.1.5 S proteins. Changes in N90A, T92I, and N322A increased infection by the early SARS-CoV-2 HU-1 strain about twofold to threefold but had lesser effects on infection by genuine Omicron variants. Despite reduced cell surface expression of ACE2, elimination of all N-linked glycosylation sites usually enhanced SARS-CoV-2 infection via the endocytic pathway while having little effect on entry at the cell surface in the presence of TMPRSS2. Our results provide insights into the role of N-linked glycans in the ability of human ACE2 (hACE2) to serve as receptors for coronavirus infection. IMPORTANCE Several human coronaviruses use angiotensin-converting enzyme 2 (ACE2) as a primary receptor for infection of human cells. ACE2 is glycosylated at seven distinct positions, and the role of glycans for the entry of SARS-CoV-2 and hCoV-NL63 into their target cells is incompletely understood. Here, we examined the impact of individual and combined mutations in hACE2 glycosylation sites on Spike-mediated VSV-pseudoparticle and genuine SARS-CoV-2 and hCoV-NL63 infection and cell-to-cell fusion. Our results provide new information on the role of glycans in hACE2 for infection by highly pathogenic and seasonal coronaviruses.
Collapse
Affiliation(s)
- Sabrina Noettger
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Rayhane Nchioua
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Chiara Pastorio
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Christoph Jung
- Institute of Electrochemistry, Ulm University, Ulm, Germany
- Electrochemical Energy Storage, Helmholtz-Institute-Ulm, Ulm, Germany
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Timo Jacob
- Institute of Electrochemistry, Ulm University, Ulm, Germany
- Electrochemical Energy Storage, Helmholtz-Institute-Ulm, Ulm, Germany
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
13
|
Razo-Blanco-Hernández DM, Hernández-Mariano JÁ, Díaz-Cureño MA, Navarrete-Martínez L, Bravata-Alcántara JC, Rivera-Sanchez R, Fernandez-Sánchez V. Association between SARS-CoV-2 viral load and serum biomarkers with mortality in Mexican patients. JOURNAL OF EDUCATION AND HEALTH PROMOTION 2025; 14:133. [PMID: 40271273 PMCID: PMC12017452 DOI: 10.4103/jehp.jehp_1481_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/05/2024] [Indexed: 04/25/2025]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has resulted in high mortality among hospitalized patients; thus, identifying mortality markers in treating these patients is essential. To evaluate the association between viral load and serum biomarkers with mortality among hospitalized patients with COVID-19. MATERIALS AND METHODS A retrospective cohort study was conducted among 198 inpatient records from a tertiary hospital in Mexico City between January and April 2021. The association between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral load and serum biomarkers with death due to COVID-19 was assessed using Cox regression models. RESULTS The median age was 54.9 years, and 61.6% were males. The mortality rate was 43.4%. After adjusting for potential confounders, patients with higher viral load [adjusted hazard ratio (aHR) = 1.56; 95% confidence interval (95% CI) = 1.01, 2.42; P value = 0.041]; and higher concentrations of BUN (aHR = 4.87;95% CI = 2.70, 8.79; P value = 0.001), creatinine (aHR = 1.60;95% CI = 1.01, 2.54; P value = 0.043), osmolality (aHR = 4.37;95% CI = 2.34, 8.14; P value = 0.001), and glucose (aHR = 2.41;95% CI = 1.40, 4.18; P value = 0.001) were more likely to have a fatal prognosis. Conversely, mortality risk was lower among patients with high concentrations of lymphocytes (aHR = 0.47;95% CI = 0.30, 0.72; P value = 0.001). CONCLUSION SARS-CoV-2 viral load and serum biomarkers such as BUN, creatinine, glucose, osmolarity, and lymphocytes could help physicians identify individuals who require closer monitoring.
Collapse
Affiliation(s)
| | | | - Mónica A. Díaz-Cureño
- Department of Medical Research and Teaching, Hospital Juárez de México, CDMX, Mexico
| | | | | | | | - Verónica Fernandez-Sánchez
- Department of Research, Hospital Juárez de México, CDMX, Mexico
- Faculty of Superior Studies Iztacala, UNAM, State of Mexico, Mexico
| |
Collapse
|
14
|
Lin D, Chen W, Lin Z, Liu L, Zhang M, Yang H, Liu Z, Chen L. Viral Transmission in Sea Food Systems: Strategies for Control and Emerging Challenges. Foods 2025; 14:1071. [PMID: 40232102 PMCID: PMC11941768 DOI: 10.3390/foods14061071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
The SARS-CoV-2 pandemic had widespread and severe impacts on both the global economy and human health. Facing the continuously mutating virus, this crisis has heightened concerns among consumers and businesses regarding viral transmission through seafood, particularly in the face of emerging, unknown viruses, underscoring our preparedness gaps. This review provides a succinct overview of the survival mechanisms of prevalent viruses in seafood, examines potential transmission pathways to humans during seafood processing, and discusses strategies for mitigating their spread throughout the seafood supply chain. Furthermore, the discussion highlights emerging trends in innovative antiviral technologies aimed at enhancing food safety. Person-to-person transmission remains the most likely source of infection within the supply chain. Therefore, it is still imperative to adhere to the implementation of standard processes, namely good manufacturing practices (GMP) and good hygiene practices (GHP), in the seafood business. In light of the significant losses caused by this crisis and the persistent presence of various viruses within the seafood supply chain, efforts are needed to implement predictive and preventive measures against potential emerging viruses. Future research should focus on monitoring and limiting viral transmission by integrating Industry 4.0 applications, smart technologies, and antiviral packaging, maximizing the potential of these emerging solutions.
Collapse
Affiliation(s)
- Dingsong Lin
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | - Wendi Chen
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | - Zejia Lin
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | - Lingdai Liu
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | - Molan Zhang
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | - Hongshun Yang
- College of Light Industry and Food Sciences, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Zifei Liu
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
| | - Lin Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| |
Collapse
|
15
|
Zhang X, Da C, Ye W. Exploratory analysis of COVID-19 propagation using logistic model. PeerJ 2025; 13:e19106. [PMID: 40124614 PMCID: PMC11927567 DOI: 10.7717/peerj.19106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
Pandemics pose significant threats to social, economic, and public health. The novel coronavirus (COVID-19), which emerged in late 2019, quickly became a global public health crisis due to its high contagion and pathogenicity. Using data from the World Health Organization (WHO), this study applied the Logistic model to analyze the spread patterns of COVID-19 in 16 countries with over 10 million infections from 2020 to 2023. The findings reveal that as of December 31, 2023, global infections exceeded 772 million with over seven million deaths. The USA and China had the highest infection numbers, while Brazil had the highest mortality rate. The study identified three main outbreak patterns: initial, late, and gradual development, reflecting different stages of the pandemic. Countries with earlier outbreaks, such as India, Brazil, the USA, and Argentina, generally had higher mortality rates, while those with later outbreaks, such as China, Pakistan, Japan, and Australia, had lower mortality rates. Significant differences were observed in the duration and speed of the spread, with China showing the shortest average duration and Russia the longest. The Logistic model's parameter k values revealed policy adjustments, with Australia, Vietnam, and China showing significant changes over time, while the USA, France, and Russia showed less impact on epidemic control. These results provide an important perspective for understanding global pandemic transmission patterns and assessing the effectiveness of quarantine strategies across countries. They also provide a scientific basis for future public health policy and pandemic response development, helping countries to develop more targeted prevention and control strategies according to the characteristics of virus transmission, rationally allocate medical resources, and reduce social harm.
Collapse
Affiliation(s)
- Xing Zhang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Chunhe Da
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Baiyin Municipal Health Commission, Baiyin, Gansu, China
- First People’s Hospital of Baiyin, Baiyin, Gansu, China
| | - Wenjuan Ye
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- First People’s Hospital of Baiyin, Baiyin, Gansu, China
| |
Collapse
|
16
|
Baissary J, Koberssy Z, Durieux JC, Atieh O, Daher J, Ailstock K, Labbato D, Foster T, Rodgers MA, Merheb A, Funderburg NT, McComsey GA. The Effect of COVID-19 on Arterial Stiffness and Inflammation: A Longitudinal Prospective Study. Viruses 2025; 17:394. [PMID: 40143322 PMCID: PMC11945347 DOI: 10.3390/v17030394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Data are limited for assessing the effect of COVID infection on endothelial function, pre- and post-pandemic. The objective of this study was to assess changes in pre-pandemic cardiovascular parameters after COVID-19 infection. This prospective cohort study used EndoPAT2000 Itamar Medical Ltd., Caesarea, Israel, to measure the augmentation index (AI; arterial elasticity) and reactive hyperemic index (RHI; endothelial function). Markers of endothelial function, inflammation, and gut integrity were collected at pre- and post-pandemic visits. COVID-negative and COVID-positive participants were matched on pre-pandemic covariates, and AI ≥ 5.0 was defined as having worse AI. Among the 156 participants, 50% had documented COVID-19 infection. Groups were balanced (p > 0.05) on pre-pandemic characteristics. Increases in oxLDL (p = 0.03) were observed in the COVID-positive group, and COVID infection had a negative effect on inflammatory markers (sVCAM-1, sTNF-RI, sTNF-RII, sCD14) and gut integrity (I-FABP, BDG) compared to COVID-negative participants (p < 0.05). There was a 16.7% (p = 0.02) increase in the proportion of COVID-positive participants with AI ≥ 5.0, without a significant change (p = 0.09) among the COVID-negative group. COVID-positive status, female sex, and higher IL-6 and sCD163 were associated (p < 0.05) with an increase in having worse AI. COVID infection is independently associated with arterial stiffness. For COVID survivors, female sex and higher markers of inflammation were associated with arterial stiffness.
Collapse
Affiliation(s)
- Jhony Baissary
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Ziad Koberssy
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Jared C. Durieux
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Ornina Atieh
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Joviane Daher
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Kate Ailstock
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; (K.A.); (N.T.F.)
| | - Danielle Labbato
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Theresa Foster
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Michael A. Rodgers
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Alexander Merheb
- Harvard Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA;
| | - Nicholas T. Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; (K.A.); (N.T.F.)
| | - Grace A. McComsey
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| |
Collapse
|
17
|
Yılmaz M, Mirzaoğlu Ç. Retrospective Cohort Study: Severe COVID-19 Leads to Permanent Blunted Heart Rate Turbulence. Diagnostics (Basel) 2025; 15:621. [PMID: 40075869 PMCID: PMC11899457 DOI: 10.3390/diagnostics15050621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Heart rate turbulence (HRT) is a non-invasive technique that can be used to evaluate autonomic nervous system (ANS) function and cardiac arrhythmia. The objective of this study is to investigate whether COVID-19 can lead to long-term blunted HRT following recovery. Methods: This retrospective cohort study included 253 individuals with a confirmed history of COVID-19, referred to as the recovered COVID-19 group, along with 315 healthy participants who had no history of the virus. The recovered COVID-19 group was categorized into three subgroups based on their chest CT severity scores. The HRT analyses were obtained from a 24-h electrocardiography-Holter recording. Results: This study revealed that the HRT onset value was elevated in the recovered COVID-19 group, while the HRT slope value showed a significant decrease when compared to the control group. Correlation analyses indicated a positive relationship between the chest CT severity score and HRT onset, whereas a negative correlation was observed between the chest CT severity score and HRT slope. Regression analyses identified recovery from severe COVID-19, chest CT severity score, hypertension (HT), and smoking as independent predictors of both abnormal HRT onset and the existence of an abnormal HRT slope. Conclusions: Individuals who have recovered from severe COVID-19 are expected to encounter a permanent blunting of HRT, which is regarded as a significant indicator of an increased risk of ventricular arrhythmias and impaired autonomic nervous system (ANS) function. Recovered severe COVID-19 individuals should be carefully evaluated for HRT with 24-h ECG-Holter.
Collapse
Affiliation(s)
- Mücahid Yılmaz
- Department of Cardiology, Elazığ Fethi Sekin City Hospital, University of Health Sciences, 23280 Elazığ, Turkey;
| | | |
Collapse
|
18
|
Jafari Z, Kolb BE, Aiken S, Wilson S. Updates on Auditory Outcomes of COVID-19 and Vaccine Side Effects: An Umbrella Review. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2025; 68:1311-1332. [PMID: 39983040 DOI: 10.1044/2024_jslhr-24-00438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
PURPOSE This umbrella review synthesizes and discusses systematic reviews (SRs) and meta-analyses (MAs) on auditory outcomes associated with COVID-19 infection and vaccination side effects. It is innovative in offering a comprehensive synthesis of evidence across adults and infants while summarizing vaccine-related auditory side effects. METHOD This literature search followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 guidelines, with no restrictions on population age or symptom severity. Four electronic databases were searched from their inception to October 2024. The Assessment of Multiple Systematic Reviews 2 checklist and Risk of Bias in Systematic Reviews tool were used to assess the quality of evidence and the risk of bias. RESULTS The systematic search identified 534 articles, narrowed down to 14 SRs following a full-text review: Nine focused on auditory outcomes of COVID-19; two, on outcomes in infants born to mothers infected during pregnancy; and three, on the auditory side effects of vaccination. A random-effects model revealed significantly high pooled estimates of hearing loss (5.0%, 95% CI [1.0, 9.0], p < .012, three MAs, N = 21,932) and tinnitus (13.5%, 95% CI [5.9, 21.1], p ≤ .001, four MAs, N = 36,236) in adults. However, current evidence in nonhospitalized patients indicates that auditory symptoms often improve after recovery. Studies also show a low rate of hearing loss in infants whose mothers contracted COVID-19 during pregnancy. Similarly, whereas COVID-19 vaccination has been linked to hearing loss and tinnitus, these effects are rare, and most patients experience improvement within weeks to months. CONCLUSIONS Evidence suggests a significantly high rate of hearing loss and tinnitus associated with COVID-19 in adults, although auditory symptoms remain rare in newborns and following vaccination. However, caution is warranted due to limitations and variability across the studies.
Collapse
Affiliation(s)
- Zahra Jafari
- School of Communication Sciences and Disorders, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Bryan E Kolb
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Alberta, Canada
| | - Steven Aiken
- School of Communication Sciences and Disorders, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sarah Wilson
- School of Communication Sciences and Disorders, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
19
|
Abou Mansour M, El Rassi C, Sleem B, Borghol R, Arabi M. Thromboembolic Events in the Era of COVID-19: A Detailed Narrative Review. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:3804576. [PMID: 40226433 PMCID: PMC11986918 DOI: 10.1155/cjid/3804576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/14/2025] [Indexed: 04/15/2025]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, is not only characterized by respiratory symptoms but is also associated with a wide range of systemic complications, including significant hematologic abnormalities. This is a comprehensive review of the current literature, using PubMed and Google Scholar, on the pathophysiology and incidence of thromboembolic events in COVID-19 patients and thromboprophylaxis. COVID-19 infection induces a prothrombotic state in patients through the dysregulation of the renin-angiotensin-aldosterone system (RAAS), endothelial dysfunction, elevated von Willebrand factor (vWF), and a dysregulated immune response involving the complement system and neutrophil extracellular traps (NETs). As a result, thromboembolic complications have emerged in COVID-19 cases, occurring more frequently in severe cases and hospitalized patients. These thrombotic events affect both venous and arterial circulation, with increased incidences of deep venous thrombosis (DVT), pulmonary embolism (PE), systemic arterial thrombosis, and myocardial infarction (MI). While DVT and PE are more common, the literature highlights the potential lethal consequences of arterial thromboembolism (ATE). This review also briefly examines the ongoing discussions regarding the use of anticoagulants for the prevention of thrombotic events in COVID-19 patients. While theoretically promising, current studies have yielded varied outcomes: Some suggest potential benefits, whereas others report an increased risk of bleeding events among hospitalized patients. Therefore, further large-scale studies are needed to assess the efficacy and safety of anticoagulants for thromboprophylaxis in COVID-19 patients.
Collapse
Affiliation(s)
- Maria Abou Mansour
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Christophe El Rassi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Bshara Sleem
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Raphah Borghol
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Hematology-Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Pediatric Department, Division of Pediatric Cardiology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
20
|
Elgarhy LH, El-Ghaiesh SH, Hamed E, Abdelfatah W. Evaluation of the safety and efficacy of isotretinoin in treatment of COVID-19 : A randomized controlled clinical trial. Contemp Clin Trials 2025; 150:107813. [PMID: 39826827 DOI: 10.1016/j.cct.2025.107813] [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: 10/23/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
The pandemic of SARS-CoV2 is not only limited to the health issues and fatalities encountered in a worldwide overwhelming burden but also the social, economic, and well-being devastation. Many trials were done to find a safe and reliable therapy for COVID-19. Isotretinoin was reported as a possible therapy for COVID-19 through the mining of post-transcriptomic and genomic datasets, which revealed isotretinoin as a potent down-regulator of the ACE2 protein the crucial gateway of SARS-CoV2 to hijack host cells. A total of 106 patients with mild to moderate COVID-19 were recruited. Patients were randomized into two groups and treated with the Standard Care (STD) protocol of the Ministry of Health, Egypt, or the STD in combination with isotretinoin (0.5 mg/kg/day) for 5 days. The study involved 66 (63 %) females and 39 (37 %) males, median age 42 years (interquartile 32-55.5 y). The main findings revealed a significant reduction in the time to improvement in the isotretinoin-treated (6.6 ± 2 d) compared to the STD-treated patients (10.4 ± 3.3). Survival analysis (HR: 4.1, 95 % CI: 2.5-6.6) in comparison to the STD-treated patients. The main adverse event reported during the therapeutic duration was the dryness of the skin, which was of acceptable tolerability through skin care instructions to the patients. The data presented herein highlights the efficacy of isotretinoin in the management of mild to moderate COVID-19 patients with a significant reduction of the time to recovery. The adverse events reported were tolerable and did not outweigh the therapeutic benefits.
Collapse
Affiliation(s)
- Lamia H Elgarhy
- Department of Dermatology, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt; Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Eman Hamed
- Department of Chest Diseases (Internal Medicine), Faculty of medicine, Suez Canal University, Ismailia, Egypt
| | - Wagdy Abdelfatah
- Department of Chest Diseases (Internal Medicine), Faculty of medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
21
|
Cappelletti G, Brambilla L, Strizzi S, Limanaqi F, Melzi V, Rizzuti M, Nizzardo M, Saulle I, Trabattoni D, Corti S, Clerici M, Biasin M. iPSC-derived human cortical organoids display profound alterations of cellular homeostasis following SARS-CoV-2 infection and Spike protein exposure. FASEB J 2025; 39:e70396. [PMID: 39950320 PMCID: PMC11826378 DOI: 10.1096/fj.202401604rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025]
Abstract
COVID-19 commonly leads to respiratory issues, yet numerous patients also exhibit a diverse range of neurological conditions, suggesting a detrimental impact of SARS-CoV-2 or the viral Spike protein on the central nervous system. Nonetheless, the molecular pathway behind neurological pathology and the presumed neurotropism of SARS-CoV-2 remains largely unexplored. We generated human cortical organoids (HCOs) derived from human induced pluripotent stem cells (hiPSC) to assess: (1) the expression of SARS-CoV-2 main entry factors; (2) their vulnerability to SARS-CoV-2 infection; and (3) the impact of SARS-CoV-2 infection and exposure to the Spike protein on their transcriptome. Results proved that (1) HCOs express the main SARS-CoV-2 receptors and co-receptors; (2) HCOs may be productively infected by SARS-CoV-2; (3) the viral particles released by SARS-CoV-2-infected HCOs are able to re-infect another cellular line; and (4) the infection resulted in the activation of apoptotic and stress pathways, along with inflammatory processes. Notably, these effects were recapitulated when HCOs were exposed to the Spike protein alone. The data obtained demonstrate that SARS-CoV-2 likely infects HCOs probably through the binding of ACE2, CD147, and NRP1 entry factors. Furthermore, exposure to the Spike protein alone proved sufficient to disrupt their homeostasis and induce neurotoxic effects, potentially contributing to the onset of long-COVID symptoms.
Collapse
Affiliation(s)
- Gioia Cappelletti
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Lorenzo Brambilla
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Sergio Strizzi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Fiona Limanaqi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Valentina Melzi
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Mafalda Rizzuti
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Monica Nizzardo
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Irma Saulle
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Daria Trabattoni
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Stefania Corti
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience SectionUniversity of MilanMilanItaly
- Neuromuscular and Rare Diseases Unit, Department of NeuroscienceFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Mario Clerici
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Don C. Gnocchi FoundationIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) FoundationMilanItaly
| | - Mara Biasin
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| |
Collapse
|
22
|
Kiratli K, Kahraman HG, Guven YZ, Akay F, Aysin M. COVID-19's effects on microvascular structure in a healthy retina: an OCTA study. Int J Ophthalmol 2025; 18:283-289. [PMID: 39967966 PMCID: PMC11754037 DOI: 10.18240/ijo.2025.02.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 10/16/2024] [Indexed: 02/20/2025] Open
Abstract
AIM To examine the subclinical alterations in the retina and choroid between patients with 2019 coronavirus disease (COVID-19)-related lung involvement and the healthy control group. METHODS In this prospective case-control study, 85 cases with lung involvement due to COVID-19 and 50 healthy cases were included. Best-corrected visual acuity, intraocular pressure measurement, and anterior and posterior segment examination were performed on both eyes for each individual. Choroidal and retinal changes were examined and recorded by optical coherence tomography angiography. RESULTS All choroidal thickness measurements of the COVID-19 group showed no statistically significant difference when compared to healthy individuals. When vascular density and perfusion density values were compared, there was a decrease in the average of these values in the COVID-19 group, although it was not statistically significant (P=0.088, P=0.065 respectively). When the fovea avascular zone (FAZ) area values were compared, the average was 0.57±0.38 in the COVID-19 group, while it was 0.54±0.24 in the control group. CONCLUSION Although our data are not statistically significant, the decrease in vascularity and perfusion and the accompanying FAZ expansion are detected in the acute period (1st month). These changes may anatomically alter the retina in the long term and affect functional vision. Future ischemia-related alterations in the retina caused by a prior COVID-19 infection may arise in situations without comorbidities and may require concern in the patient's systemic assessment.
Collapse
Affiliation(s)
- Kazim Kiratli
- Department of Infectious Diseases and Clinical Microbiology, Katip Celebi University Ataturk Educating and Research Hospital, Izmir 35360, Türkiye
| | - Hazan Gul Kahraman
- Department of Ophthalmology, Democracy University Buca Seyfi Demirsoy Educating and Research Hospital, Izmir 35390, Türkiye
| | - Yusuf Ziya Guven
- Department of Ophthalmology, Katip Çelebi University Atatürk Educating and Research Hospital, Izmir 35360, Türkey
| | - Fahrettin Akay
- Department of Ophthalmology, Health Sciences University Gülhane Educating and Research Hospital, Ankara 06010, Türkiye
| | - Murat Aysin
- Department of Public Health, Balikesir University Faculty of Medicine, Balikesir 10230, Türkiye
| |
Collapse
|
23
|
Quinn AE, Zhao L, Bell SD, Huq MH, Fang Y. Exploring Asthma as a Protective Factor in COVID-19 Outcomes. Int J Mol Sci 2025; 26:1678. [PMID: 40004141 PMCID: PMC11855143 DOI: 10.3390/ijms26041678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Asthma has long been associated with increased susceptibility to viral respiratory infections, leading to significant exacerbations and poorer clinical outcomes. Contrarily and interestingly, emerging data and research surrounding the COVID-19 pandemic have shown that patients with asthma infected with SARS-CoV-2 experienced decreased severity of disease, lower hospitalization rates, as well as decreased morbidity and mortality. Research has shown that eosinophils could enhance immune defense against viral infections, while inhaled corticosteroids can assist in controlling systematic inflammation. Moreover, reduced ACE-2 expression in individuals with asthma may restrict viral entry, and the Th2 immune response may offset the Th1 response typically observed in severe COVID-19 patients. These factors may help explain the favorable outcomes seen in asthmatic patients during the COVID-19 pandemic. This review highlights potential protective mechanisms seen in asthmatic patients, including eosinophilia, the use of inhaled corticosteroids, reduced ACE-2 expression, and a dominate Th2 immune response. Such a study will be helpful to better manage patients with asthma who have contracted COVID-19.
Collapse
Affiliation(s)
- Anthony E. Quinn
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Muhammad H. Huq
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
24
|
Luo YW, Huang AL, Tang KF. Angiotensin-converting enzyme 2 and hepatic SARS-CoV-2 infection: Regulation, association, and therapeutic implications. World J Gastroenterol 2025; 31:100864. [PMID: 39958440 PMCID: PMC11752700 DOI: 10.3748/wjg.v31.i6.100864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/07/2024] [Accepted: 12/20/2024] [Indexed: 01/10/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters host cells via the angiotensin-converting enzyme 2 (ACE2) receptor. Mounting evidence has indicated the presence of hepatic SARS-CoV-2 infection and liver injury in patients with coronavirus disease 2019 (COVID-19). Understanding the mechanisms of hepatic SARS-CoV-2 infection is crucial for addressing COVID-19-related liver pathology and developing targeted therapies. This editorial discusses the significance of ACE2 in hepatic SARS-CoV-2 infection, drawing on the research by Jacobs et al. Their findings indicate that hepatic ACE2 expression, frequency of hepatic SARS-CoV-2 infection, and severity of liver injury are elevated in patients with pre-existing chronic liver diseases. These data suggest that hepatic ACE2 could be a promising therapeutic target for COVID-19.
Collapse
Affiliation(s)
- Yu-Wei Luo
- Key Laboratory of Molecular Biology on Infectious Disease, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Ai-Long Huang
- Key Laboratory of Molecular Biology on Infectious Disease, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Kai-Fu Tang
- Key Laboratory of Molecular Biology on Infectious Disease, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
25
|
Spiteri S, Salamon I, Girolamini L, Pascale MR, Marino F, Derelitto C, Caligaris L, Paghera S, Ferracin M, Cristino S. Surfaces environmental monitoring of SARS-CoV-2: Loop mediated isothermal amplification (LAMP) and droplet digital PCR (ddPCR) in comparison with standard Reverse-Transcription quantitative polymerase chain reaction (RT-qPCR) techniques. PLoS One 2025; 20:e0317228. [PMID: 39899502 PMCID: PMC11790120 DOI: 10.1371/journal.pone.0317228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
The persistence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) on substrates, and the impact of fomites on Coronavirus Disease 19 (COVID-19) transmission, is until now, widely discussed. Consequently, further investigations are required for a correct risk assessment in high-risk facilities such as hospitals, healthcare facilities (HCFs), and long-term care facilities (LTCFs). Therefore, appropriate surveillance and disinfection programs represent the best approach to guarantee the safety of these communities. This study proposes an environmental SARS-CoV-2 surfaces routine monitoring approach in HCF and communities' settings, to provide rapid and effective evaluation of surface hygienic conditions and the effectiveness of applied sanitization measures. Surfaces samples (n = 118) were collected using the SRK® kit (Copan Italia) from 2020 to 2023. Three molecular techniques were compared: Reverse Transcription Loop mediated isothermal AMPlification (RT-LAMP, Enbiotech), Reverse-Transcription quantitative polymerase chain reaction (RT-qPCR) (RT-qPCR, Seegene) and droplet digital PCR (ddPCR, Bio-Rad). For ddPCR, two RNA extraction methods were compared: TRIzol LS (Invitrogen) versus QIAmp Viral Mini kit (QIAGEN), showing how the latter is more suitable for surfaces. Regarding the quantitative ddPCR results, the ROC analysis allowed to reduce the manufacturer cut-off for droplets number (from 3 to 1) for the positive samples. Moreover, a new cut-off for the viral RNA copies' number/μL for each target (N1 and N2) on environmental monitoring was fixed at 2,82. The results obtained using the QIAmp kit, suggested that the N2 target is more stable in the environment and could be most suitable for the virus environmental detection. The percentage of positive samples was similar among the techniques (26% for RT-LAMP, 36% for ddPCR and 23% for RT-qPCR). Using RT-qPCR as reference method, a sensitivity (SE) of 30% for RT-LAMP and 41% for ddPCR was observed. By contrast, specificity (SP) was higher for RT-LAMP (75%) respect to ddPCR (66%). Comparing the faster RT-LAMP with the sensitive ddPCR the 26% and 74% of SE and SP for RT-LAMP, were reported. The low sensitivity for RT-LAMP and ddPCR could be explained with the use of clinical rather than environmental kits, other than the changing in the virus prevalence during the sampling campaign. Although the RT-LAMP requires improvements in term of SE and SP, this research presents an innovative environmental monitoring and prevention method for SARS-CoV-2, that could be extended to other pathogens that are under environmental surveillance.
Collapse
Affiliation(s)
- Simona Spiteri
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Irene Salamon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luna Girolamini
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Maria Rosaria Pascale
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Federica Marino
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Carlo Derelitto
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Laura Caligaris
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | | | - Manuela Ferracin
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Sandra Cristino
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Batlle D, Hassler L, Wysocki J. ACE2, From the Kidney to SARS-CoV-2: Donald Seldin Award Lecture 2023. Hypertension 2025; 82:166-180. [PMID: 39624896 DOI: 10.1161/hypertensionaha.124.22064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
ACE2 (angiotensin-converting enzyme 2) is a monocarboxypeptidase that cleaves Ang II (angiotensin II) among other substrates. ACE2 is present in the cell membrane of many organs, most abundantly in epithelial cells of kidney proximal tubules and the small intestine, and also exists in soluble forms in plasma and body fluids. Membrane-bound ACE2 exerts a renoprotective action by metabolizing Ang II and therefore attenuating the undesirable actions of excess Ang II. Therefore, soluble ACE2, by downregulating this peptide, may exert a therapeutic action. Our laboratory has designed ACE2 truncates that pass the glomerular filtration barrier to target the kidney renin-angiotensin system directly and, therefore, compensate for loss of kidney membrane-bound ACE2. Membrane-bound ACE2 is also the essential receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Soluble ACE2 proteins have been studied as a way to intercept SARS-CoV-2 from binding to membrane-bound ACE2 and prevent cell entry of SARS-CoV-2 altogether. We bioengineered a soluble ACE2 protein, termed ACE2 618-DDC-ABD, with increased binding affinity for SARS-CoV-2 and prolonged duration of action, which, when administered intranasally, provides near-complete protection from lethality in k18hACE2 mice infected with different SARS-CoV-2 variants. The main advantage of soluble ACE2 proteins for the neutralization of SARS-CoV-2 is their immediate onset of action and universality for current and future emerging SARS-CoV-2 variants. It is notable that ACE2 is critically involved in 2 dissimilar functions: as a receptor for cell entry of many coronaviruses and as an enzyme in the metabolism of Ang II, and yet in both cases, it is a therapeutic target.
Collapse
Affiliation(s)
- Daniel Batlle
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Luise Hassler
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jan Wysocki
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
27
|
Kochi M, Yokoyama Y. [Posterior reversible encephalopathy syndrome after COVID-19 in a patient with chronic renal failure]. Rinsho Shinkeigaku 2025; 65:32-38. [PMID: 39710393 DOI: 10.5692/clinicalneurol.cn-002013] [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] [Indexed: 12/24/2024]
Abstract
A 61-year-old man with chronic renal failure had an embolic stroke of undetermined source that was treated with warfarin. Five weeks later, the patient contracted coronavirus disease (COVID-19). Six days after the onset of COVID-19, high blood pressure (>200 mmHg) and consciousness disturbance were reported. CT demonstrated symmetrical hypodensity areas in the bilateral cerebellar hemispheres. MRI revealed hyperintensity lesions in the bilateral cerebellar hemispheres and pons on the T2-weighted and fluid-attenuated inversion recovery images. Moreover, cerebellar lesions appeared as hyperintensity areas on apparent diffusion coefficient mapping. Based on these findings, a diagnosis of posterior reversible encephalopathy syndrome (PRES) was made. The patient was treated with antihypertensive drugs, and the consciousness level improved gradually. MRI after one month showed that the lesions had disappeared. PRES should be considered if the brain CT of patients with COVID-19 shows a low-density lesion, especially in patients with risk factors for PRES such as chronic renal failure or hypertension.
Collapse
|
28
|
Han Y, Guo J, Li X, Zhong Z. Differences in clinical characteristics between coronavirus disease 2019 (COVID-19) and influenza: a systematic review and meta-analysis. NPJ Prim Care Respir Med 2025; 35:8. [PMID: 39875405 PMCID: PMC11775258 DOI: 10.1038/s41533-025-00414-0] [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: 06/27/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) epidemic has brought major challenges to the global health system, and influenza is also a problem that cannot be ignored. We aimed to explore and compare the clinical characteristics of COVID-19 and influenza to deepen the understanding of these two diseases and provide some guidance for clinicians to make differential diagnoses. We searched PubMed, Embase and Web of Science for articles and performed a meta-analysis using Stata 14.0 with a random-effects model. This meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. One hundred articles involving 226,913 COVID-19 patients and 201,617 influenza patients were included, and all the articles included patients with these two diseases as experimental and control groups. Compared to influenza, COVID-19 was more common among men (OR = 1.46, 95% CI: 1.23-1.74) and people with a higher body mass index (MD = 1.43, 95% CI: 1.09-1.77). The proportion of current smokers among COVID-19 patients was lower than that among influenza patients (OR = 0.25, 95% CI: 0.18-0.33). Patients with COVID-19 had longer stays in the hospital (MD = 3.20, 95% CI: 2.58-3.82) and ICU (MD = 3.10, 95% CI: 1.44-4.76), required mechanical ventilation more frequently (OR = 2.30, 95% CI: 1.77-3.00), and had higher mortality (OR = 2.22, 95% CI: 1.93-2.55). We also found significant differences in some blood parameters between the two groups of patients. Upper respiratory symptoms were more obvious in influenza patients, and the proportion of comorbidities was higher than that among COVID-19 patients. There are some differences in the major characteristics, symptoms, laboratory findings and comorbidities between COVID-19 patients and influenza patients. COVID-19 patients often require more medical resources and have worse clinical outcomes.
Collapse
Affiliation(s)
- Yingying Han
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jia Guo
- Department of Respiratory, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xingzhao Li
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhuan Zhong
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
29
|
Sharma G, Kumar N, Sharma CS, Alqahtani T, Tiruneh YK, Sultana S, Rolim Silva GV, de Lima Menezes G, Zaki MEA, Nobre Oliveira JI. Identification of promising SARS-CoV-2 main protease inhibitor through molecular docking, dynamics simulation, and ADMET analysis. Sci Rep 2025; 15:2830. [PMID: 39843610 PMCID: PMC11754916 DOI: 10.1038/s41598-025-86016-9] [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: 09/12/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 continues to pose a major challenge to global health. Targeting the main protease of the virus (Mpro), which is essential for viral replication and transcription, offers a promising approach for therapeutic intervention. In this study, advanced computational techniques such as molecular docking and molecular dynamics simulations were used to screen a series of antiviral compounds for their potential inhibitory effect on the SARS-CoV-2 Mpro. A comprehensive analysis of compounds from the ChemDiv and PubChem databases was performed. The physicochemical properties, pharmacokinetics, and ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) profiles were evaluated to determine drug similarity and safety. Compound 4896 - 4038 proved to be the most promising candidate. It exhibited a favorable balance between molecular weight (491.06) and lipophilicity (logP 3.957), high intestinal absorption (92.119%), and broad tissue distribution (VDss of 0.529), indicating good oral bioavailability and therapeutic potential. Molecular docking studies showed that 4896 - 4038 has a strong binding affinity to the active site of Mpro and forms key interactions, such as hydrogen bonds, carbon-hydrogen bonds, pi-sulfur, and multiple van der Waals and pi-pi stacked bonds. The binding energy was comparable to that of the reference drug X77, indicating potential efficacy. Molecular dynamics simulations over 300 ns confirmed the stability of the Mpro/4896 - 4038 complex of protein-ligand. Free energy landscape mapping and MM/PBSA calculations further substantiated the favorable binding and stability of the complex. Importantly, 4896 - 4038 exhibited a comparatively favorable safety profile. In summary, compound 4896 - 4038 shows significant potential as a potent SARS-CoV-2 Mpro inhibitor, combining potent inhibitory activity with favorable pharmacokinetic and safety profiles. These results support the further development of 4896 - 4038 as a promising therapeutic agent in the fight against COVID-19 that warrants experimental validation and clinical investigation.
Collapse
Affiliation(s)
- Ganesh Sharma
- Department of Pharmaceutical Chemistry, Bhupal Nobles' College of Pharmacy, Bhupal Nobles' University, Udaipur, 313002, India.
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles' College of Pharmacy, Bhupal Nobles' University, Udaipur, 313002, India
| | - Chandra Shekhar Sharma
- Department of Pharmaceutical Chemistry, Bhupal Nobles' College of Pharmacy, Bhupal Nobles' University, Udaipur, 313002, India
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Yewulsew Kebede Tiruneh
- Department: Biology, Biomedical Sciences stream Bahir Dar University, Bahir Dar, P.O.Box=79, Bahir Dar, Ethiopia.
| | - Sharifa Sultana
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, 59064-741, RN, Brazil
| | - Gabriel Vinícius Rolim Silva
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, 59064-741, RN, Brazil
| | - Gabriela de Lima Menezes
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, 59064-741, RN, Brazil
| | - Magdi E A Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia.
| | - Jonas Ivan Nobre Oliveira
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, 59064-741, RN, Brazil.
| |
Collapse
|
30
|
Xiao F, Hu J, Xu M, Wang D, Shen X, Zhang H, Miao J, Cai H, Wang J, Liu Y, Xiao S, Zhu L. Animal Models for Human-Pathogenic Coronavirus and Animal Coronavirus Research. Viruses 2025; 17:100. [PMID: 39861889 PMCID: PMC11768759 DOI: 10.3390/v17010100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Coronavirus epidemics have posed a serious threat to both human and animal health. To combat emerging infectious diseases caused by coronaviruses, various animal infection models have been developed and applied in research, including non-human primate models, ferret models, hamster models, mouse models, and others. Moreover, new approaches have been utilized to develop animal models that are more susceptible to infection. These approaches include using viral delivery methods to induce the expression of viral receptors in mouse tissues and employing gene-editing techniques to create genetically modified mice. This has led to the successful establishment of infection models for multiple coronaviruses, significantly advancing related research. In contrast, livestock and pets that can be infected by animal coronaviruses provide valuable insights when used as infection models, enabling the collection of accurate clinical data through the analysis of post-infection pathological features. However, despite the potential insights, there is a paucity of research data pertaining to these infection models. In this review, we provide a detailed overview of recent progress in the development of animal models for coronaviruses that cause diseases in both humans and animals and suggest ways in which animal models can be adapted to further enhance their value in research.
Collapse
Affiliation(s)
- Fenglian Xiao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
- Traditional Chinese Medicine and Health School, Nanfang College, Guangzhou 510970, China
| | - Jincheng Hu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Minsheng Xu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Di Wang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Xiaoyan Shen
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Hua Zhang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Jie Miao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Haodong Cai
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Jihui Wang
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Yaqing Liu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Shan Xiao
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
| | - Longchao Zhu
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China; (F.X.); (J.H.); (M.X.); (D.W.); (X.S.); (H.Z.); (J.M.); (H.C.); (J.W.); (Y.L.)
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
31
|
Alicia LB, María Ángeles OG, Desirée MG, Maximino R, Marilina GA. Utility of Protein Markers in COVID-19 Patients. Int J Mol Sci 2025; 26:653. [PMID: 39859366 PMCID: PMC11766239 DOI: 10.3390/ijms26020653] [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: 12/12/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
COVID-19 has been a challenge at the healthcare level not only in the early stages of the pandemic, but also in the subsequent appearance of long-term COVID-19. Several investigations have attempted to identify proteomic biomarkers in an attempt to improve clinical care, guide treatment and predict possible patient outcomes. Proteins such as C-reactive protein (CRP) or interleukin 6 (IL-6) are clear markers of severe disease, but many others have been proposed that could help in risk stratification and in the prediction of specific complications. This review aims to bring together the most relevant studies in this regard, providing information to identify the most notable biomarkers in relation to COVID-19 found to date.
Collapse
Affiliation(s)
- López-Biedma Alicia
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
| | - Onieva-García María Ángeles
- Preventive Medicine and Public Health Unit, Hospital Universitario Reina Sofia, 14004 Cordoba, Spain;
- Preventive Medicine and Public Health Research Group, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain
| | - Martín-García Desirée
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - Redondo Maximino
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - García-Aranda Marilina
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
| |
Collapse
|
32
|
Spalinger MR, Sanati G, Chatterjee P, Hai R, Li J, Santos AN, Nordgren TM, Tremblay ML, Eckmann L, Hanson E, Scharl M, Wu X, Boland BS, McCole DF. Tofacitinib Mitigates the Increased SARS-CoV-2 Infection Susceptibility Caused by an IBD Risk Variant in the PTPN2 Gene. Cell Mol Gastroenterol Hepatol 2025; 19:101447. [PMID: 39756517 PMCID: PMC11953972 DOI: 10.1016/j.jcmgh.2024.101447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND & AIMS Coronavirus disease (COVID-19), caused by severe acquired respiratory syndrome-Coronavirus-2 (SARS-CoV-2), triggered a global pandemic with severe medical and socioeconomic consequences. Although fatality rates are higher among the elderly and those with underlying comorbidities, host factors that promote susceptibility to SARS-CoV-2 infection and severe disease are poorly understood. Although individuals with certain autoimmune/inflammatory disorders show increased susceptibility to viral infections, there is incomplete knowledge of SARS-CoV-2 susceptibility in these diseases. The aim of our study was to investigate whether the autoimmunity risk gene, PTPN2, which also confers elevated risk to develop inflammatory bowel disease, affects susceptibility to SARS-CoV-2 viral uptake. METHODS Using samples from PTPN2 genotyped patients with inflammatory bowel disease, PTPN2-deficient mice, and human intestinal and lung epithelial cell lines, we investigated how PTPN2 affects expression of the SARS-CoV-2 receptor angiotensin converting enzyme 2 (ACE2), and uptake of virus-like particles expressing the SARS-CoV2 spike protein and live SARS-CoV-2 virus. RESULTS We report that the autoimmune PTPN2 loss-of-function risk variant rs1893217 promotes expression of the SARS-CoV-2 receptor, ACE2, and increases cellular entry of SARS-CoV-2 spike protein and live virus. Elevated ACE2 expression and viral entry were mediated by increased Janus kinase-signal transducers and activators of transcription signaling and were reversed by the Janus kinase inhibitor, tofacitinib. CONCLUSION Collectively, our findings uncover a novel risk biomarker for increased expression of the SARS-CoV-2 receptor and viral entry, and identify a clinically approved therapeutic agent to mitigate this risk.
Collapse
Affiliation(s)
- Marianne R Spalinger
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California; Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Golshid Sanati
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Pritha Chatterjee
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Rong Hai
- Department of Microbiology and Plant Pathology, University of California Riverside, Riverside, California
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Alina N Santos
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California; Current position: College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Michel L Tremblay
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Lars Eckmann
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Elaine Hanson
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Monrovia, California
| | - Brigid S Boland
- Division of Gastroenterology, University of California San Diego, La Jolla, California
| | - Declan F McCole
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California.
| |
Collapse
|
33
|
Di Domenico M, Motta A, Dai Pra T, Cantore S, Dioguardi M, Zanella ER, Arrigoni R, De Vito D, Mastrangelo F. The "Magnificent Seven" in Oral and Systemic Health against COVID-19. Endocr Metab Immune Disord Drug Targets 2025; 25:271-280. [PMID: 38798209 DOI: 10.2174/0118715303296892240506100532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024]
Abstract
The COVID-19 pandemic impacted all areas of daily life, including medical care. Unfortunately, to date, no specific treatments have been found for the cure of this disease, and therefore, it is advisable to implement all possible strategies to prevent infection. In this context, it is important to better define the role of all behaviors, in particular nutrition, in order to establish whether these can both prevent infection and improve the outcome of the disease in patients with COVID-19. There is sufficient evidence to demonstrate that immune response can be weakened by inadequate nutrition. Nutrition management and treatment are very important to enhance the immune response of an infected person against RNA viral infection. A complete nutritional assessment should include anthropometric, dietary, and laboratorial assessment, as well as a multidisciplinary discussion about the patient's clinical condition. In this way, it is possible to establish an individualized nutritional approach to contribute to improving clinical and nutritional prognoses. From this point of view, diet, through intake of vitamins and trace elements and maintaining adequate functioning of the intestinal barrier, can reduce the severity of the COVID-19 infection. In this study, we provide an overview of the effects of diet on COVID-19 infection in non-cancer patients. This notion needs to be further evaluated, and thus, identification, characterization, and targeting of the right nutrition principles related to the management of patients with COVID-19 are likely to improve outcomes and may prevent the infection or lead to a cure.
Collapse
Affiliation(s)
- Marina Di Domenico
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessandro Motta
- Operative Unit of Oral Surgery for Special Needs and Dentistry, Trento Hospital, Borgo Valsugana, Trento, Italy
| | - Tommaso Dai Pra
- Operative Unit of Oral Surgery for Special Needs and Dentistry, Trento Hospital, Borgo Valsugana, Trento, Italy
| | - Stefania Cantore
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Dioguardi
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | | | - Roberto Arrigoni
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
- School of Medicine, University of Bari, Aldo Moro, Bari, Italy
| | - Danila De Vito
- School of Medicine, University of Bari, Aldo Moro, Bari, Italy
| | - Filiberto Mastrangelo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
34
|
Blanco J, Trinité B, Puig‐Barberà J. Rethinking Optimal Immunogens to Face SARS-CoV-2 Evolution Through Vaccination. Influenza Other Respir Viruses 2025; 19:e70076. [PMID: 39871737 PMCID: PMC11773156 DOI: 10.1111/irv.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/23/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
SARS-CoV-2, which originated in China in late 2019, quickly fueled the global COVID-19 pandemic, profoundly impacting health and the economy worldwide. A series of vaccines, mostly based on the full SARS-CoV-2 Spike protein, were rapidly developed, showing excellent humoral and cellular responses and high efficacy against both symptomatic infection and severe disease. However, viral evolution and the waning humoral neutralizing responses strongly challenged vaccine long term effectiveness, mainly against symptomatic infection, making necessary a strategy of repeated and updated booster shots. In this repeated vaccination context, antibody repertoire diversification was evidenced, although immune imprinting after booster doses or reinfection was also demonstrated and identified as a major determinant of immunological responses to repeated antigen exposures. Considering that a small domain of the SARS-CoV-2 Spike protein, the receptor binding domain (RBD), is the major target of neutralizing antibodies and concentrates most viral mutations, the following text aims to provide insights into the ongoing debate over the best strategies for vaccine boosters. We address the relevance of developing new booster vaccines that target the evolving RBD, thus focusing on the relevant antigenic sites of the SARS-CoV-2 new variants. A combination of this strategy with immunofusing and computerized approaches could minimize immune imprinting, therefore optimizing neutralizing immune responses and booster vaccine efficacy.
Collapse
Affiliation(s)
- Julià Blanco
- IrsiCaixaBadalonaCataloniaSpain
- Germans Trias i Pujol Research Institute (IGTP)BadalonaCataloniaSpain
- CIBER de Enfermedades InfecciosasMadridSpain
- Chair in Infectious Diseases and Immunity, Faculty of MedicineUniversity of Vic‐Central University of Catalonia (UVic‐UCC)VicCataloniaSpain
| | | | - Joan Puig‐Barberà
- Área de Investigación en VacunasFundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat ValencianaValenciaSpain
| |
Collapse
|
35
|
Bai R, Liang B, Guo Y, Liu W, Wen Z, Wang Z, Zhang Y, Du J, Song Y, Yu Z, Ma X. ACE2 Inhibits Dermal Regeneration Through Ang II in Tissue Expansion. J Cosmet Dermatol 2025; 24:e16767. [PMID: 39829293 PMCID: PMC11744341 DOI: 10.1111/jocd.16767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/02/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Tissue expansion is a widely employed technique in reconstructive surgery aimed at addressing considerable skin defects. Nevertheless, matters like inadequate expansion capability and the potential for skin breakage due to the fragility of the expanded tissue present notable hurdles in enhancing skin regeneration during this process. Angiotensin-converting enzyme 2 (ACE2) is recognized for its essential role in facilitating tissue renewal and regeneration. However, its precise impact on skin renewal during tissue expansion remains underexplored. This study seeks to elucidate ACE2's contribution to skin regeneration, specifically examining its role in collagen synthesis. METHODS This study evaluated the expression and distribution of ACE2 in expanded skin using samples derived from both rats and human patients. Additionally, we investigated ACE2 expression in stretched keratinocytes in vitro. ACE2 knockout keratinocytes were transfected with small interfering RNA (siRNA) and cocultured with fibroblasts to observe fibroblast proliferation and migration. MLN-4760 was utilized to inhibit the ACE2 enzymatic activity. Additionally, we analyzed parameters such as the size of expanded skin, dermal thickness, and the levels of collagen I (COL I), collagen III (COL III), and transforming growth factor β (TGF-β) to elucidate the role of ACE2 in the context of expanded skin. RESULTS The thinning of the expanded dermis was linked with elevated ACE2 expression. Enzymatic activity and ACE2 expression were both increased by mechanical stress. Additionally, ACE2 utilized Ang II to activate the migration and proliferation of human dermal fibroblasts. In vivo, the ACE2 inhibitor MLN-4760 promoted skin regeneration and reduced dermal thinning by elevating COL I, COL III, and TGF-β during expansion. CONCLUSIONS This finding suggest that mechanical stretch increases ACE2 expression, which in turn promotes the regeneration of expanded skin. The basis for using ACE2 in clinical settings to increase tissue expansion efficacy is provided by this work.
Collapse
Affiliation(s)
- Ruoxue Bai
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Baoyan Liang
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Yaotao Guo
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Wei Liu
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Zhuoyue Wen
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Zhantong Wang
- Xijing 986 Hospital DepartmentFourth Military Medical UniversityXi'anShaanxiChina
| | - Yu Zhang
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Jing Du
- Key Laboratory of Aerospace Medicine of Ministry of EducationSchool of Aerospace Medicine, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Yajuan Song
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Zhou Yu
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| | - Xianjie Ma
- Department of Plastic and Reconstructive SurgeryXijing Hospital, Fourth Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
36
|
Castillo-Galán S, Parra V, Cuenca J. Unraveling the pathogenesis of viral-induced pulmonary arterial hypertension: Possible new therapeutic avenues with mesenchymal stromal cells and their derivatives. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167519. [PMID: 39332781 DOI: 10.1016/j.bbadis.2024.167519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Pulmonary hypertension (PH) is a severe condition characterized by elevated pressure in the pulmonary artery, where metabolic and mitochondrial dysfunction may contribute to its progression. Within the PH spectrum, pulmonary arterial hypertension (PAH) stands out with its primary pulmonary vasculopathy. PAH's prevalence varies from 0.4 to 1.4 per 100,000 individuals and is associated with diverse conditions, including viral infections such as HIV. Notably, recent observations highlight an increased occurrence of PAH among COVID-19 patients, even in the absence of pre-existing cardiopulmonary disorders. While current treatments offer partial relief, there's a pressing need for innovative therapeutic strategies, among which mesenchymal stromal cells (MSCs) and their derivatives hold promise. This review critically evaluates recent investigations into viral-induced PAH, encompassing pathogens like human immunodeficiency virus, herpesvirus, Cytomegalovirus, Hepatitis B and C viruses, SARS-CoV-2, and Human endogenous retrovirus K (HERKV), with a specific emphasis on mitochondrial dysfunction. Furthermore, we explore the underlying rationale driving novel therapeutic modalities, including MSCs, extracellular vesicles, and mitochondrial interventions, within the framework of PAH management.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Valentina Parra
- Laboratory of Differentiation and Cell Metabolism (D&M), Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; SYSTEMIX Center for Systems Biology, O'Higgins University, Rancagua, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile; Cells for Cells, Santiago, Chile.
| |
Collapse
|
37
|
Behzadi P, Chandran D, Chakraborty C, Bhattacharya M, Saikumar G, Dhama K, Chakraborty A, Mukherjee S, Sarshar M. The dual role of toll-like receptors in COVID-19: Balancing protective immunity and immunopathogenesis. Int J Biol Macromol 2025; 284:137836. [PMID: 39613064 DOI: 10.1016/j.ijbiomac.2024.137836] [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: 06/09/2024] [Revised: 11/01/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Toll-like receptors (TLRs) of human are considered as the most critical immunological mediators of inflammatory pathogenesis of COVID-19. These immunoregulatory glycoproteins are located on the surface and/or intracellular compartment act as innate immune sensors. Upon binding with distinct SARS-CoV-2 ligand(s), TLRs signal activation of different transcription factors that induce expression of the proinflammatory mediators that collectively induce 'cytokine storm'. Similarly, TLR activation is also pivotal in conferring protection to infection and invasion as well as upregulating the tissue repair pathways. This dual role of the human TLRs in deciding the fate of SARS-CoV-2 has made these receptor proteins as the critical mediators of immunoprotective and immunopathogenic consequences associated with COVID-19. Herein, pathbreaking discoveries exploring the immunobiological importance of the TLRs in COVID-19 and developing TLR-directed therapeutic intervention have been reviewed by accessing the up-to-date literatures available in the public domain/databases. In accordance with our knowledge in association with the importance of TLRs' role against viruses and identification of viral particles, they have been recognized as suitable candidates with high potential as vaccine adjuvants. In this regard, the agonists of TLR4 and TLR9 have effective potential in vaccine technology while the others need further investigations. This comprehensive review suggests that basal level expression of TLRs can act as friends to keep our body safe from strangers but act as a foe via overexpression. Therefore, selective inhibition of the overexpressed TLRs appears to be a solution to counteract the cytokine storm while TLR-agonists as vaccine adjuvants could lessen the risk of infection in the naïve population.
Collapse
Affiliation(s)
- Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, 37541-374, Iran.
| | | | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, VyasaVihar, Balasore, 756020, Odisha, India
| | - Guttula Saikumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India.
| | - Ankita Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India.
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, 00146, Rome, Italy
| |
Collapse
|
38
|
Upadhyay PK, Thakur N, Vishwakarma VK, Chaurasiya HS, Ansari TM. Modulation of Angiotensin-II and Angiotensin 1-7 Levels Influences Cardiac Function in Myocardial Ischemia-reperfusion Injury. Curr Drug Res Rev 2025; 17:102-112. [PMID: 38299413 DOI: 10.2174/0125899775280160240122065607] [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: 09/26/2023] [Revised: 12/11/2023] [Accepted: 01/16/2024] [Indexed: 02/02/2024]
Abstract
The angiotensin-converting enzyme-2 (ACE-2) alters the pathophysiology of various fatal cardiovascular diseases, including ischemic heart disease, whereas angiotensin 1-7 (Ang 1-7) exerts a wide range of actions. The effects of ischemia-reperfusion (IR) injury include damage to myocardial tissue that initiates protease action, causing cardiac cell death. Angiotensin- II (Ang-II) contributes through the renin-angiotensin system (RAS) to the IR injury, whereas Ang 1-7 paradoxically exerts a protective effect through the same. Thus, the myocardial ischemic reperfusion injury (MIRI) may be altered by the RAS of the heart. This review paper focuses on ACE-2, angiotensin-converting enzyme (ACE), and Ang 1-7 regulation in the RAS of the heart in the pathophysiology of MIRI. The treatment in such conditions using ACE-2 activator, ACE inhibitor, and Ang-II antagonists may promote vascular functions as well as cardio- protection.
Collapse
Affiliation(s)
- Prabhat Kumar Upadhyay
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| | - Navneet Thakur
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | |
Collapse
|
39
|
Ko C, Cheng CC, Mistretta D, Ambike S, Sacherl J, Velkov S, Liao BH, Bester R, Gültan M, Polezhaeva O, Herrmann A, Jakwerth CA, Schmidt-Weber CB, Bugert JJ, Wölfel R, Grass V, Essbauer S, Schnepf D, Keppler OT, Vondran FWR, Pichlmair A, Mogler C, Ebert G, Protzer U. SARS-CoV-2 Productively Infects Human Hepatocytes and Induces Cell Death. J Med Virol 2025; 97:e70156. [PMID: 39760326 DOI: 10.1002/jmv.70156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
SARS-CoV-2 infection is accompanied by elevated liver enzymes, and patients with pre-existing liver conditions experience more severe disease. While it was known that SARS-CoV-2 infects human hepatocytes, our study determines the mechanism of infection, demonstrates viral replication and spread, and highlights direct hepatocyte damage. Viral replication was readily detectable upon infection of primary human hepatocytes and hepatoma cells with the ancestral SARS-CoV-2, Delta, and Omicron variants. Hepatocytes express the SARS-CoV-2 receptor ACE2 and the host cell protease TMPRSS2, and knocking down ACE2 and TMPRSS2 impaired SARS-CoV-2 infection. Progeny viruses released from infected hepatocytes showed the typical coronavirus morphology by electron microscopy and proved infectious when transferred to fresh cells, indicating that hepatocytes can contribute to virus spread. Importantly, SARS-CoV-2 infection rapidly induced hepatocyte death in a replication-dependent fashion, with the Omicron variant showing faster onset but less extensive cell death. C57BL/6 wild-type mice infected with a mouse-adapted SARS-CoV-2 strain showed high levels of viral RNA in liver and lung tissues. ALT peaked when viral RNA was cleared from the liver. Liver histology revealed profound tissue damage and immune cell infiltration, indicating that direct cytopathic effects of SARS-CoV-2 and immune-mediated killing of infected hepatocytes contribute to liver pathology.
Collapse
Grants
- This study was supported by the German Research Foundation (DFG) via SFB-TRR179 (project 272983813 to U.P.), TRR22 (project 398577603 to C.S.W.) and TRR353 (project 471011418 to G.E.), by the State of Bavaria via research network FOR-COVID and Bay-VOC, by the project "Virological and immunological determinants of COVID-19 pathogenesis-lessons to get prepared for future pandemics" (KA1-Co-02 "COVIPA" to U.P.) and "Airborne Transmission of SARS Coronavirus - From Fundamental Science to Efficient Air Cleaning Systems" (KA1-Co-06 "CORAERO" to G.E.), grants from the Helmholtz Association's Initiative and Networking Fund, by the European Commission FET Open Grant VIROFIGHT (grant no. 899619), by the State of Bavaria and the European Union via a grant for regional infrastructure development (EFRE - REACT, to U.P. and G.E.), by the State of Bavaria via research networks FOR-COVID and Bay-VOC (to U.P. and O.T.K.) by the Federal Ministry of Education and Research (project ESCAPE; 01KI20169A to C.S.W.), and by the Medical Biological Defense Research Program of the Bundeswehr Medical Service (to J.J.B.). In addition, this research was supported by intramural funds from KRICT (project KK2432-10 and BSF24-111 to C.K.).
Collapse
Affiliation(s)
- Chunkyu Ko
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, South Korea
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Daniele Mistretta
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Shubhankar Ambike
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Julia Sacherl
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Stoyan Velkov
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Romina Bester
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Merve Gültan
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Olga Polezhaeva
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Alexander Herrmann
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich/Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich/Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Lung Research (DZL), Munich Partner Site, Munich, Germany
| | - Joachim J Bugert
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Roman Wölfel
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Vincent Grass
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Sandra Essbauer
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Oliver T Keppler
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
- Max von Pettenkofer Institute & Gene Center, Faculty of Medicine, University of Munich, Munich, Germany
| | - Florian W R Vondran
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
- ReMediES, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| |
Collapse
|
40
|
Güven O, Karakurt G, Naser A, Selçuk H, Keleş DV, Gedik E, Avsever M, Köse FF. The Impact of COVID-19 Infection on the Development of Stroke, Pulmonary Embolism, and Myocardial Infarction: A Retrospective Study. Cureus 2025; 17:e77665. [PMID: 39968441 PMCID: PMC11835032 DOI: 10.7759/cureus.77665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
INTRODUCTION This study compares the period during which thromboembolic disease develops after contact with the virus before, during, and after the pandemic. METHODS In this study, the medical records of patients with a preliminary diagnosis of myocardial infarction (MI), pulmonary embolism (PE), and ischemic stroke who presented to the Emergency Department before, during, and after the pandemic (when vaccination rates increased) were retrospectively examined. Data on whether these patients had COVID-19 or were vaccinated, the time interval between infection/vaccination and the onset of these conditions, and the prognosis were analyzed. RESULTS In the MI group, patients developed embolism the longest after infection and the shortest after vaccination. Among MI patients, the rate of those who received the BioNTech vaccine during the normalization period was higher than that of those who received Sinovac (p = 0.005). In stroke patients, during the pandemic, the time to post-vaccine embolism was shorter (p < 0.001). Additionally, infection and vaccination increased the mortality rate in stroke and PE patients (p < 0.001). CONCLUSION This study demonstrates that thromboembolic events can occur at varying rates and durations after exposure to the virus. While the causes of thrombosis are multifactorial, contact with the virus may act as a triggering factor, even if COVID-19 does not have a direct effect.
Collapse
Affiliation(s)
- Oya Güven
- Department of Emergency Medicine, Kırklareli University Faculty of Medicine, Kırklareli, TUR
- Department of Emergency Medicine, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Gökhan Karakurt
- Department of Pulmonology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Abdulrahman Naser
- Department of Cardiology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Hakan Selçuk
- Department of Emergency Medicine, Babaeskı State Hospital, Kırklareli, TUR
| | - Dilek V Keleş
- Department of Nursing, Kırklareli University Faculty of Health Sciences, Kırklareli, TUR
| | - Emre Gedik
- Department of Neurology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Mert Avsever
- Department of Emergency Medicine, St. John's Hospital, Edinburgh, GBR
| | - Fatih Furkan Köse
- Department of Emergency Medicine, Kırklareli Training and Research Hospital, Kırklareli, TUR
| |
Collapse
|
41
|
Grelewska‐Nowotko K, Elhag AE, Turowski TW. Transcription Kinetics in the Coronavirus Life Cycle. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70000. [PMID: 39757745 PMCID: PMC11701415 DOI: 10.1002/wrna.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025]
Abstract
Coronaviruses utilize a positive-sense single-strand RNA, functioning simultaneously as mRNA and the genome. An RNA-dependent RNA polymerase (RdRP) plays a dual role in transcribing genes and replicating the genome, making RdRP a critical target in therapies against coronaviruses. This review explores recent advancements in understanding the coronavirus transcription machinery, discusses it within virus infection context, and incorporates kinetic considerations on RdRP activity. We also address steric limitations in coronavirus replication, particularly during early infection phases, and outline hypothesis regarding translation-transcription conflicts, postulating the existence of mechanisms that resolve these issues. In cells infected by coronaviruses, abundant structural proteins are synthesized from subgenomic RNA fragments (sgRNAs) produced via discontinuous transcription. During elongation, RdRP can skip large sections of the viral genome, resulting in the creation of shorter sgRNAs that reflects the stoichiometry of viral structural proteins. Although the precise mechanism of discontinuous transcription remains unknown, we discuss recent hypotheses involving long-distance RNA-RNA interactions, helicase-mediated RdRP backtracking, dissociation and reassociation of RdRP, and RdRP dimerization.
Collapse
Affiliation(s)
| | - Ahmed Eisa Elhag
- Institute of Biochemistry and BiophysicsPolish Academy of SciencesWarsawPoland
- Department of Preventive Medicine and Clinical Studies, Faculty of Veterinary SciencesUniversity of GadarifAl QadarifSudan
| | | |
Collapse
|
42
|
Becker RC, Tantry US, Khan M, Gurbel PA. The COVID-19 thrombus: distinguishing pathological, mechanistic, and phenotypic features and management. J Thromb Thrombolysis 2025; 58:15-49. [PMID: 39179952 PMCID: PMC11762605 DOI: 10.1007/s11239-024-03028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 08/26/2024]
Abstract
A heightened risk for thrombosis is a hallmark of COVID-19. Expansive clinical experience and medical literature have characterized small (micro) and large (macro) vessel involvement of the venous and arterial circulatory systems. Most events occur in patients with serious or critical illness in the hyperacute (first 1-2 weeks) or acute phases (2-4 weeks) of SARS-CoV-2 infection. However, thrombosis involving the venous, arterial, and microcirculatory systems has been reported in the subacute (4-8 weeks), convalescent (> 8-12 weeks) and chronic phases (> 12 weeks) among patients with mild-to-moderate illness. The purpose of the current focused review is to highlight the distinguishing clinical features, pathological components, and potential mechanisms of venous, arterial, and microvascular thrombosis in patients with COVID-19. The overarching objective is to better understand the proclivity for thrombosis, laying a solid foundation for screening and surveillance modalities, preventive strategies, and optimal patient management.
Collapse
Affiliation(s)
- Richard C Becker
- Cardiovascular Center, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA.
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| | - Muhammad Khan
- Division of General Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Baltimore, USA
| |
Collapse
|
43
|
Changela S, Ashraf S, Lu JY, Duong KE, Henry S, Wang SH, Duong TQ. New-onset gastrointestinal disorders in COVID-19 patients 3.5 years post-infection in the inner-city population in the Bronx. Sci Rep 2024; 14:31850. [PMID: 39738536 PMCID: PMC11685902 DOI: 10.1038/s41598-024-83232-7] [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: 07/22/2024] [Accepted: 12/12/2024] [Indexed: 01/02/2025] Open
Abstract
This study examined the incidence, characteristics, and risk factors of new gastrointestinal disorders (GID) associated with SARS-CoV-2 infection up to 3.5 years post-infection. This retrospective study included 35,102 COVID-19 patients and 682,594 contemporary non-COVID-19 patients without past medical history of GID (controls) from the Montefiore Health System in the Bronx (3/1/2020 to 7/31/2023). Comparisons were made with unmatched and propensity-matched (1:2) controls. The primary outcome was new GID which included peptic ulcer, inflammatory bowel disease, irritable bowel syndrome, diverticulosis, diverticulitis, and biliary disease. Multivariate Cox proportional hazards model analysis was performed with adjustment for covariates. There were 2,228 (6.34%) COVID-19 positive patients who developed new GID compared to 38,928 (5.70%) controls. COVID-19 patients had an elevated risk of developing new GID (adjusted HR = 1.18 (95% CI 1.12-1.25) compared to propensity-matched controls, after adjusting for confounders that included smoking, obesity, diabetes, hypertension. These findings underscore the need for additional research and follow-up of at-risk individuals for developing GID post infection.
Collapse
Affiliation(s)
- Sagar Changela
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Samad Ashraf
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Justin Y Lu
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Kevin E Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Sonya Henry
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Stephen H Wang
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Tim Q Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Center for Health & Data Innovation, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
44
|
Angius F, Puxeddu S, Zaimi S, Canton S, Nematollahzadeh S, Pibiri A, Delogu I, Alvisi G, Moi ML, Manzin A. SARS-CoV-2 Evolution: Implications for Diagnosis, Treatment, Vaccine Effectiveness and Development. Vaccines (Basel) 2024; 13:17. [PMID: 39852796 PMCID: PMC11769326 DOI: 10.3390/vaccines13010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, driven by the rapid evolution of the SARS-CoV-2 virus, presents ongoing challenges to global public health. SARS-CoV-2 is characterized by rapidly evolving mutations, especially in (but not limited to) the spike protein, complicating predictions about its evolutionary trajectory. These mutations have significantly affected transmissibility, immune evasion, and vaccine efficacy, leading to multiple pandemic waves with over half a billion cases and seven million deaths globally. Despite several strategies, from rapid vaccine development and administration to the design and availability of antivirals, including monoclonal antibodies, already having been employed, the persistent circulation of the virus and the emergence of new variants continue to result in high case numbers and fatalities. In the past four years, immense research efforts have contributed much to our understanding of the viral pathogenesis mechanism, the COVID-19 syndrome, and the host-microbe interactions, leading to the development of effective vaccines, diagnostic tools, and treatments. The focus of this review is to provide a comprehensive analysis of the functional impact of mutations on diagnosis, treatments, and vaccine effectiveness. We further discuss vaccine safety in pregnancy and the implications of hybrid immunity on long-term protection against infection, as well as the latest developments on a pan-coronavirus vaccine and nasal formulations, emphasizing the need for continued surveillance, research, and adaptive public health strategies in response to the ongoing SARS-CoV-2 evolution race.
Collapse
Affiliation(s)
- Fabrizio Angius
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvia Puxeddu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvio Zaimi
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Serena Canton
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Sepehr Nematollahzadeh
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Andrea Pibiri
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Ilenia Delogu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| |
Collapse
|
45
|
Tiniakou E, Casciola‐Rosen L, Thomas MA, Manabe Y, Antar AAR, Damarla M, Hassoun PM, Gao L, Wang Z, Zeger S, Rosen A. Autoantibodies in hospitalised patients with COVID-19. Clin Transl Immunology 2024; 13:e70019. [PMID: 39734590 PMCID: PMC11671454 DOI: 10.1002/cti2.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/31/2024] Open
Abstract
Objectives CD209L and its homologous protein CD209 act as alternative entry receptors for the SARS-CoV-2 virus and are highly expressed in the virally targeted tissues. We tested for the presence and clinical features of autoantibodies targeting these receptors and compared these with autoantibodies known to be associated with COVID-19. Methods Using banked samples (n = 118) from Johns Hopkins patients hospitalised with COVID-19, we defined autoantibodies against CD209 and CD209L by enzyme-linked immunosorbent assay (ELISA). Clinical associations of these antibodies were compared with those of patients with anti-interferon (IFN) and anti-angiotensin-converting enzyme-2 (ACE2) autoantibodies. Results Amongst patients hospitalised with COVID-19, 19.5% (23/118) had IgM autoantibodies against CD209L and were more likely to have coronary artery disease (44% vs 19%, P = 0.03). Antibodies against CD209 were present in 5.9% (7/118); interestingly, all 7 were male (P = 0.02). In our study, the presence of either antibody was positively associated with disease severity [OR 95% confidence interval (95% CI): 1.80 (0.69-5.03)], but the association did not reach statistical significance. In contrast, 10/118 (8.5%) had IgG autoantibodies against IFNα, and 21 (17.8%) had IgM antibodies against ACE2. These patients had significantly worse prognosis (intubation or death) and prolonged hospital stays. However, when adjusting for patient characteristics on admission, only the presence of anti-ACE2 IgM remained significant [pooled common OR (95% CI), 4.14 (1.37, 12.54)]. Conclusion We describe IgM autoantibodies against CD209 and CD209L amongst patients hospitalised with COVID-19. These were not associated with disease severity. Conversely, patients with either anti-ACE2 IgM or anti-IFNα IgG antibodies had worse outcomes. Due to the small size of the study cohort, conclusions drawn should be considered cautiously.
Collapse
Affiliation(s)
- Eleni Tiniakou
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Livia Casciola‐Rosen
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mekha A Thomas
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Yuka Manabe
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Annukka AR Antar
- Division of Infectious Diseases, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Li Gao
- Division of Allergy and Immunology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreMDUSA
| | - Zitong Wang
- Department of BiostatisticsBloomberg School of Public HealthBaltimoreMDUSA
| | - Scott Zeger
- Department of BiostatisticsBloomberg School of Public HealthBaltimoreMDUSA
| | - Antony Rosen
- Division of Rheumatology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
46
|
Yu S, Song C. Clinical characteristics of coronavirus disease 2019 patients with hepatitis B virus super-infection. Rev Inst Med Trop Sao Paulo 2024; 66:e74. [PMID: 39699512 DOI: 10.1590/s1678-9946202466074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/29/2024] [Indexed: 12/20/2024] Open
Abstract
COVID-19 and hepatitis B disease are significant global pandemics, both of which can lead to liver damage. This study aims to report the clinical course of liver function and disease prognosis of COVID-19 patients with hepatitis B virus (HBV) super-infections. A total of 249 outpatients with COVID-19 were enrolled in this study from December 1, 2023 to February 28, 2024. Clinical characteristics, laboratory data, chest CT findings, and patients' treatment and outcomes were collected and analyzed retrospectively. Of the 249 outpatients, 37 (14.9%) were super-infected with HBV, whereas 212 (85.1%) showed no such outcome. This study found no significant differences between the two groups regarding age, gender, symptoms, complications, or chest CT findings. However, COVID-19 patients super-infected with HBV showed lower white blood cell, neutrophil, and platelet counts (p < 0.05). Additionally, total bilirubin levels were significantly higher in the SARS-CoV-2/HBV super-infected group compared to the COVID-19-only group (p = 0.022). After the first week of similar treatment, both groups showed almost identical outcomes, including hospitalization, severity, and mortality rates. Thus, SARS-CoV-2/HBV super-infection slightly affected liver function but did not worsen COVID-19 outcomes. Routine HBV monitoring and liver function tests are recommended to manage COVID-19 patients with HBV super-infections. This study found no clear indications of the need to change the therapeutic prescription for COVID-19 in cases of HBV super-infections.
Collapse
Affiliation(s)
- Shan Yu
- Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of Infectious Diseases, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shuguang Hospital, Shanghai, China
| | - Cunzheng Song
- Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital, Department of Infectious Diseases, Shanghai, China
| |
Collapse
|
47
|
Chiavarini M, Dolcini J, Firmani G, Ponzio E, Barbadoro P. Prevalence of Diabetes, Hypertension, and Associated of Cardiovascular Diseases: A Comparative Pre- and Post-COVID Study. Diseases 2024; 12:329. [PMID: 39727659 PMCID: PMC11727044 DOI: 10.3390/diseases12120329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
Background: Diabetes and hypertension are major global health challenges aggravated by COVID-19's impact on healthcare and lifestyle factors. This study aims to compare the prevalence and associated socio-demographic factors of these conditions before and after the pandemic (2019 vs. 2022). Materials and Methods: We used data from Italy's "Aspects of Daily Life" survey; 74,294 adults were included. Results: Results show a rise in diabetes prevalence from 7.76% in 2019 to 8.49% in 2022 (p < 0.05), while hypertension did not show this. Logistic regression analysis for the years 2019 and 2022 revealed a statistically significant association between the year 2022 and increased odds of diabetes (OR = 1.08, p = 0.008). BMI's role as a risk factor intensified, with higher odds ratios (ORs) for both conditions in overweight and obese individuals in 2022. For example, obesity-related ORs for diabetes increased from 2.45 (95%CI 1.73-3.47) in 2019 to 3.02 (95%CI 2.09-4.35) in 2022, and for hypertension from 2.86 (95%CI 2.28-3.58) to 3.64 (95%CI 2.87-4.61). Lower education levels also showed a greater association with hypertension risk in 2022; subjects with only middle or high school diplomas had significantly higher ORs than individuals with higher education; there was a non-significant trend in 2019. However, diabetes risk associated with lower education remained stable and significant in both years. Conclusions: These findings suggest that the pandemic may have increased risk factors for diabetes and hypertension, particularly BMI and educational level, compared with the literature on the increased burden of chronic diseases during COVID-19.
Collapse
Affiliation(s)
- Manuela Chiavarini
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy
- Department of Health Sciences, University of Florence, Viale GB Morgagni 48, 50134 Florence, Italy
| | - Jacopo Dolcini
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy
| | - Giorgio Firmani
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy
| | - Elisa Ponzio
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy
| | - Pamela Barbadoro
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, 60126 Ancona, Italy
| |
Collapse
|
48
|
Maghsoodi E, Hasanpour E, Soleimani F, Aghal M, Mollarahimi K. Evaluation of anxiety level and the factors Affecting Anxiety in health care workers in Shahid Dr. Gholipour Hospital, Bukan, Iran during COVID-19 pandemic. PLoS One 2024; 19:e0264916. [PMID: 39671554 PMCID: PMC11644838 DOI: 10.1371/journal.pone.0264916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/21/2022] [Indexed: 12/15/2024] Open
Abstract
INTRODUCTION The rapid and dangerous spread of Covid-19 has caused psychological problems, especially anxiety among health care workers. Due to the lack of accurate information on the prevalence of anxiety and the factors affecting it among health care workers, especially in developing countries, this study aimed to investigate the level of anxiety and the effective factors in health care workers in Shahid Dr. Gholipour Hospital in Bukan, Iran in Covid-19 pandemic. METHODOLOGY A descriptive cross-sectional study was carried out through census sampling among health care workers of the hospital. Totally, 358 participants entered the study. Data gathering tool consisted of a demographical information tool and Spielberger's Anxiety scale. To determine the effective variables on hidden and obvious anxiety use liner regression. significant predictors variable of anxiety was determined by logistic regression. FINDINGS Means score for obvious anxiety was 47.005 (5.4) CI: 46.47-47.54 and for hidden anxiety was 42.790 (4.88) CI: 42.43-43.29. Factors affecting participants' obvious anxiety based on the Unadjusted model were Work experience, Age group, Marital status and History of anxiety disorders and gender. In the Adjusted model, the age group and the anxiety history are considered as effective variables. One of the effective factors on hidden anxiety is the effect of gender variable in both Adjusted and Unadjusted models. Based on logistic regression test, job variables, age groups, marital status (are predicted variables of obvious anxiety. In hidden anxiety, marital status was introduced as a predictor variable. CONCLUSION The majority of personnel experienced a high level of anxiety during the pandemic. Psychological examination and interventions are essential for the health care workers.
Collapse
Affiliation(s)
- Esmaiel Maghsoodi
- Department of Nursing, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Edris Hasanpour
- Department of Nursing. Boukan faculty of Nursing, Urmia University of Medical Sciences, Urmia, Iran
| | - Farzaneh Soleimani
- Department of Nursing, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Moosa Aghal
- Department of Nursing, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Keyvan Mollarahimi
- Department of Nursing. Boukan faculty of Nursing, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
49
|
Matviichuk A, Yerokhovych V, Zemskov S, Ilkiv Y, Gurianov V, Shaienko Z, Falalyeyeva T, Sulaieva O, Kobyliak N. Unveiling risk factors for post-COVID-19 syndrome development in people with type 2 diabetes. Front Endocrinol (Lausanne) 2024; 15:1459171. [PMID: 39722811 PMCID: PMC11668646 DOI: 10.3389/fendo.2024.1459171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Post-COVID-19 syndrome (PCS) is a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection-associated chronic condition characterized by long-term violations of physical and mental health. People with type 2 diabetes (T2D) are at high risk for severe COVID-19 and PCS. Aim The current study aimed to define the predictors of PCS development in people with T2D for further planning of preventive measures and improving patient outcomes. Materials and methods The data were collected through the national survey targeting persons with T2D concerning the history of COVID-19 course and signs and symptoms that developed during or after COVID-19 and continued for more than 12 weeks and were not explained by an alternative diagnosis. In total, 469 patients from different regions of Ukraine were enrolled in the study. Among them, 227 patients reported PCS development (main group), while 242 patients did not claim PCS symptoms (comparison group). Stepwise multivariate logistic regression and probabilistic neural network (PNN) models were used to select independent risk factors. Results Based on the survey data, 8 independent factors associated with the risk of PCS development in T2D patients were selected: newly diagnosed T2D (OR 4.86; 95% CI 2.55-9.28; p<0.001), female sex (OR 1.29; 95% CI 0.86-1.94; p=0.220), COVID-19 severity (OR 1.35 95% CI 1.05-1.70; p=0.018), myocardial infarction (OR 2.42 95% CI 1.26-4.64; p=0.002) and stroke (OR 3.68 95% CI 1.70-7.96; p=0.001) in anamnesis, HbA1c above 9.2% (OR 2.17 95% CI 1.37-3.43; p=0.001), and the use of insulin analogs (OR 2.28 95% CI 1.31-3.94; p=0.003) vs human insulin (OR 0.67 95% CI 0.39-1.15; p=0.146). Although obesity aggravated COVID-19 severity, it did not impact PCS development. In ROC analysis, the 8-factor multilayer perceptron (MLP) model exhibited better performance (AUC 0.808; 95% CІ 0.770-0.843), allowing the prediction of the risk of PCS development with a sensitivity of 71.4%, specificity of 76%, PPV of 73.6% and NPV of 73.9%. Conclusions Patients who were newly diagnosed with T2D, had HbA1c above 9.2%, had previous cardiovascular or cerebrovascular events, and had severe COVID-19 associated with mechanical lung ventilation were at high risk for PCS.
Collapse
Affiliation(s)
- Anton Matviichuk
- Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine
| | | | - Sergii Zemskov
- Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Yeva Ilkiv
- Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Vitalii Gurianov
- Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Zlatoslava Shaienko
- Department of Endocrinology with Pediatric Infectious Diseases, Poltava State Medical University, Poltava, Ukraine
| | - Tetyana Falalyeyeva
- Department of Fundamental Medicine, Educational-Scientific Center “Institute of Biology and Medicine” Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
- Scientific Department, Medical Laboratory CSD, Kyiv, Ukraine
| | - Oksana Sulaieva
- Scientific Department, Medical Laboratory CSD, Kyiv, Ukraine
- Department of Pathology, Kyiv Medical University, Kyiv, Ukraine
| | - Nazarii Kobyliak
- Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine
- Scientific Department, Medical Laboratory CSD, Kyiv, Ukraine
| |
Collapse
|
50
|
Rong Z, Mai H, Ebert G, Kapoor S, Puelles VG, Czogalla J, Hu S, Su J, Prtvar D, Singh I, Schädler J, Delbridge C, Steinke H, Frenzel H, Schmidt K, Braun C, Bruch G, Ruf V, Ali M, Sühs KW, Nemati M, Hopfner F, Ulukaya S, Jeridi D, Mistretta D, Caliskan ÖS, Wettengel JM, Cherif F, Kolabas ZI, Molbay M, Horvath I, Zhao S, Krahmer N, Yildirim AÖ, Ussar S, Herms J, Huber TB, Tahirovic S, Schwarzmaier SM, Plesnila N, Höglinger G, Ondruschka B, Bechmann I, Protzer U, Elsner M, Bhatia HS, Hellal F, Ertürk A. Persistence of spike protein at the skull-meninges-brain axis may contribute to the neurological sequelae of COVID-19. Cell Host Microbe 2024; 32:2112-2130.e10. [PMID: 39615487 DOI: 10.1016/j.chom.2024.11.007] [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: 08/26/2024] [Revised: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024]
Abstract
SARS-CoV-2 infection is associated with long-lasting neurological symptoms, although the underlying mechanisms remain unclear. Using optical clearing and imaging, we observed the accumulation of SARS-CoV-2 spike protein in the skull-meninges-brain axis of human COVID-19 patients, persisting long after viral clearance. Further, biomarkers of neurodegeneration were elevated in the cerebrospinal fluid from long COVID patients, and proteomic analysis of human skull, meninges, and brain samples revealed dysregulated inflammatory pathways and neurodegeneration-associated changes. Similar distribution patterns of the spike protein were observed in SARS-CoV-2-infected mice. Injection of spike protein alone was sufficient to induce neuroinflammation, proteome changes in the skull-meninges-brain axis, anxiety-like behavior, and exacerbated outcomes in mouse models of stroke and traumatic brain injury. Vaccination reduced but did not eliminate spike protein accumulation after infection in mice. Our findings suggest persistent spike protein at the brain borders may contribute to lasting neurological sequelae of COVID-19.
Collapse
Affiliation(s)
- Zhouyi Rong
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Medical Research School (MMRS), Munich, Germany
| | - Hongcheng Mai
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Munich Medical Research School (MMRS), Munich, Germany
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Saketh Kapoor
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Czogalla
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Senbin Hu
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jinpeng Su
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Danilo Prtvar
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Inderjeet Singh
- Research Unit Adipocytes & Metabolism (ADM), Helmholtz Diabetes Center, Helmholtz Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claire Delbridge
- Institute of Pathology, Division of Neuropathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hanno Steinke
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Hannah Frenzel
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Katja Schmidt
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Christian Braun
- Institute of Legal Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Gina Bruch
- Institute of Legal Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Viktoria Ruf
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mayar Ali
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Graduate School of Neuroscience (GSN), Munich, Germany
| | | | - Mojtaba Nemati
- Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Franziska Hopfner
- Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany
| | - Denise Jeridi
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany
| | - Daniele Mistretta
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | | | | | - Fatma Cherif
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Graduate School of Neuroscience (GSN), Munich, Germany
| | - Müge Molbay
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Medical Research School (MMRS), Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Center of Doctoral Studies in Informatics and its Applications (CEDOSIA), Technical University of Munich, Munich, Germany
| | - Shan Zhao
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Siegfried Ussar
- Research Unit Adipocytes & Metabolism (ADM), Helmholtz Diabetes Center, Helmholtz Munich, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Susanne M Schwarzmaier
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Benjamin Ondruschka
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Markus Elsner
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany
| | - Harsharan Singh Bhatia
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Farida Hellal
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Munich, Neuherberg, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Koç University, School of Medicine, İstanbul, Turkey.
| |
Collapse
|