1
|
Wang J, Xiong Y, Song Z, Li Y, Zhang L, Qin C. Progress in research on osteoporosis secondary to SARS-CoV-2 infection. Animal Model Exp Med 2025; 8:829-841. [PMID: 40029778 DOI: 10.1002/ame2.12573] [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: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 05/28/2025] Open
Abstract
The World Health Organization has declared that COVID-19 no longer constitutes a "public health emergency of international concern," yet the long-term impact of SARS-CoV-2 infection on bone health continues to pose new challenges for global public health. In recent years, numerous animal model and clinical studies have revealed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to secondary osteoporosis. The mechanisms involved are related to the virus's direct effects on bone tissue, dysregulation of the body's inflammatory response, hypoxia, noncoding RNA imbalance, and metabolic abnormalities. Although these studies have unveiled the connection between SARS-CoV-2 infection and osteoporosis, current research is not comprehensive and in depth. Future studies are needed to evaluate the long-term effects of SARS-CoV-2 on bone density and metabolism, elucidate the specific mechanisms of pathogenesis, and explore potential interventions. This review aims to collate existing research literature on SARS-CoV-2 infection-induced secondary osteoporosis, summarize the underlying mechanisms, and provide direction for future research.
Collapse
Affiliation(s)
- Jinlong Wang
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
- Changping National Laboratory (CPNL), Beijing, China
| | - Yibai Xiong
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Zhiqi Song
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Yanhong Li
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Ling Zhang
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Chuan Qin
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
- Changping National Laboratory (CPNL), Beijing, China
| |
Collapse
|
2
|
Fry LG, Washam CL, Roys H, Bowlin AK, Venugopal G, Bird JT, Byrum SD, Weinkopff T. HIF-α signaling regulates the macrophage inflammatory response during Leishmania major infection. Front Immunol 2025; 16:1487311. [PMID: 40191198 PMCID: PMC11969800 DOI: 10.3389/fimmu.2025.1487311] [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: 08/27/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Cutaneous leishmaniasis (CL) contributes significantly to the global burden of neglected tropical diseases, with 12 million people currently infected with Leishmania parasites. CL encompasses a range of disease manifestations, from self-healing skin lesions to permanent disfigurations. Currently there is no vaccine available, and many patients are refractory to treatment, emphasizing the need for new therapeutic targets. Previous work demonstrated macrophage HIF-α-mediated lymphangiogenesis is necessary to achieve efficient wound resolution during murine L. major infection. Here, we investigate the role of macrophage HIF-α signaling independent of lymphangiogenesis. We sought to determine the relative contributions of the parasite and the host-mediated inflammation in the lesional microenvironment to myeloid HIF-α signaling. Because HIF-α activation can be detected in infected and bystander macrophages in leishmanial lesions, we hypothesize it is the host's inflammatory response and microenvironment, rather than the parasite, that triggers HIF-α activation. To address this, macrophages from mice with intact HIF-α signaling (LysMCreARNTf/+) or mice with deleted HIF-α signaling (LysMCreARNTf/f) were subjected to RNASequencing after L. major infection and under pro-inflammatory stimulus. We report that L. major infection alone is enough to induce some minor HIF-α-dependent transcriptomic changes, while infection with L. major in combination with pro-inflammatory stimuli induces numerous transcriptomic changes that are both dependent and independent of HIF-α signaling. Additionally, by coupling transcriptomic analysis with several pathway analyses, we found HIF-α suppresses pathways involved in protein translation during L. major infection in a pro-inflammatory environment. Together these findings show L. major induces a HIF-α-dependent transcriptomic program, but HIF-α only suppresses protein translation in a pro-inflammatory environment. Thus, this work indicates the host inflammatory response, rather than the parasite, largely contributes to myeloid HIF-α signaling during Leishmania infection.
Collapse
Affiliation(s)
- Lucy G. Fry
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Anne K. Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jordan T. Bird
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
3
|
D’Amico RC, Nagashima S, Carstens LB, Bertoldi KDG, Mataruco S, Honório D’Agostini JC, Hlatchuk EC, da Silva SB, de Noronha L, Baena CP. COVID-19 Induces Greater NLRP3 Inflammasome Activation in Obese Patients than Other Chronic Illnesses: A Case-Control Study. Int J Mol Sci 2025; 26:1541. [PMID: 40004007 PMCID: PMC11855377 DOI: 10.3390/ijms26041541] [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: 12/23/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity has been identified as an independent risk factor for severe COVID-19 unfavorable outcomes. Several factors, such as increased ACE2 receptor expression and chronic inflammation, can contribute to this relationship, yet the activation of the NLRP3 inflammasome pathway is also a key element. Our primary goal was to determine whether chronic NLRP3 inflammasome activation in people with obesity is different in critical COVID-19 and in critical chronic conditions. A retrospective analysis was conducted using clinical data and post-mortem lung tissue samples from 14 COVID-19 patients with obesity (group A) and 9 patients with obesity who died from non-COVID-19 causes (group B). Immunohistochemical analysis assessed twelve markers related to the NLRP3 inflammasome pathway. Group A showed a significantly higher expression of ASC (p = 0.0387) and CASP-1 (p = 0.0142). No significant differences were found for IL-8, TNF-α, NF-kB, NLRP3, IL-1β, and gasdermin-D. Group B had higher levels of IL-6 (p < 0.0001), IL-18 (p = 0.002), CASP-9 (p < 0.0001), and HIF (p = 0.0327). We concluded that COVID-19 activates the NLRP3 inflammasome pathway, possibly leading to pyroptotic cell death mediated by caspase-1. In contrast, people with obesity without COVID-19, despite exhibiting some markers of the NLRP3 inflammasome, are more likely to experience necroptosis mediated by caspase-9.
Collapse
Affiliation(s)
- Raíssa Campos D’Amico
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Seigo Nagashima
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Lucas Baena Carstens
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Karina de Guadalupe Bertoldi
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Sabrina Mataruco
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | | | - Elisa Carolina Hlatchuk
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Sofia Brunoro da Silva
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Lucia de Noronha
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Cristina Pellegrino Baena
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| |
Collapse
|
4
|
Al-Kuraishy HM, Al-Gareeb AI, Al-Maiahy TJ, Alexiou A, Mukerjee N, Batiha GES. An insight into the placental growth factor (PlGf)/angii axis in Covid-19: a detrimental intersection. Biotechnol Genet Eng Rev 2024; 40:3326-3345. [PMID: 36096720 DOI: 10.1080/02648725.2022.2122291] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/29/2022] [Indexed: 11/02/2022]
Abstract
Coronavirus disease 2019 (Covid-19) is a recent and current infectious pandemic caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). Covid-19 may lead to the development of acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and extrapulmonary manifestations in severe cases. Down-regulation of angiotensin-converting enzyme (ACE2) by the SARS-CoV-2 increases the production of angiotensin II (AngII), which increases the release of pro-inflammatory cytokines and placental growth factor (PlGF). PlGF is a critical molecule involved in vasculogenesis and angiogenesis. PlGF is stimulated by AngII in different inflammatory diseases through a variety of signaling pathways. PlGF and AngII are interacted in SARS-CoV-2 infection resulting in the production of pro-inflammatory cytokines and the development of Covid-19 complications. Both AngII and PlGF are interacted and are involved in the progression of inflammatory disorders; therefore, we aimed in this review to highlight the potential role of the PlGF/AngII axis in Covid-19.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Thabat J Al-Maiahy
- Department Of Gynecology and Obstetrics, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
- AFNP Med, Austria, Wien, Austria
| | - Nobendu Mukerjee
- Department of Microbiology; Ramakrishna Mission Vivekananda Centenary College, Kolkata, WestBengal, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, Egypt
| |
Collapse
|
5
|
Fehsel K. Metabolic Side Effects from Antipsychotic Treatment with Clozapine Linked to Aryl Hydrocarbon Receptor (AhR) Activation. Biomedicines 2024; 12:2294. [PMID: 39457607 PMCID: PMC11505606 DOI: 10.3390/biomedicines12102294] [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: 05/31/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic syndrome (MetS) is the most common adverse drug reaction from psychiatric pharmacotherapy. Neuroreceptor blockade by the antipsychotic drug clozapine induces MetS in about 30% of patients. Similar to insulin resistance, clozapine impedes Akt kinase activation, leading to intracellular glucose and glutathione depletion. Additional cystine shortage triggers tryptophan degradation to kynurenine, which is a well-known AhR ligand. Ligand-bound AhR downregulates the intracellular iron pool, thereby increasing the risk of mitochondrial dysfunction. Scavenging iron stabilizes the transcription factor HIF-1, which shifts the metabolism toward transient glycolysis. Furthermore, the AhR inhibits AMPK activation, leading to obesity and liver steatosis. Increasing glucose uptake by AMPK activation prevents dyslipidemia and liver damage and, therefore, reduces the risk of MetS. In line with the in vitro results, feeding experiments with rats revealed a disturbed glucose-/lipid-/iron-metabolism from clozapine treatment with hyperglycemia and hepatic iron deposits in female rats and steatosis and anemia in male animals. Decreased energy expenditure from clozapine treatment seems to be the cause of the fast weight gain in the first weeks of treatment. In patients, this weight gain due to neuroleptic treatment correlates with an improvement in psychotic syndromes and can even be used to anticipate the therapeutic effect of the treatment.
Collapse
Affiliation(s)
- Karin Fehsel
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Bergische Landstrasse 2, 40629 Duesseldorf, Germany
| |
Collapse
|
6
|
Zhang J, Kondowe B, Zhang H, Xie X, Song Q, Guo B, Shang J. Identification of prognostic indicator based on hypoxia-related lncRNAs analysis in lung adenocarcinoma. Malawi Med J 2024; 36:170-178. [PMID: 40018402 PMCID: PMC11862855 DOI: 10.4314/mmj.v36i3.3] [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] [Indexed: 03/01/2025] Open
Abstract
Introduction There were no systematic studies about hypoxia-related long noncoding RNAs (lncRNAs) signatures to predict the survival of patients with lung adenocarcinoma (LUAD). Setting up matching hypoxia-related lncRNA signatures was necessary. Objective This study aimed to establish hypoxia-related lncRNAs signatures and to seek new biomarkers to predict the prognosis of the patients with lung adenocarcinoma. Methodology The Cancer Genome Atlas (TCGA) database provided the expression profiles of lncRNAs that includes 535 lung adenocarcinoma samples. The coexpression network of lncRNAs and hypoxia-related different expression genes (DEGs) was utilized to select hypoxia-related lncRNAs. The lncRNAs were further screened using univariate Cox regression. In addition, Lasso regression and multivariate Cox regression were used to develop a hypoxia-related lncRNAs signature. A risk score based on the signature was established, and Cox regression was used to test if it was an independent prognostic factor. Results Nine prognostic hypoxia-related lncRNAs (LINC01150, AC010980.2, AL606489.1, AL034397.3, LINC00460, LINC02081, FAM83AAS1, AL365181.2, and AC026355.1) were identified to be significantly different, which made up a hypoxia-related lncRNAs signature. The high-risk group had shorter OS compared with the low-risk group (P = 3.329e - 09, log-rank test). A risk score based on the signature was a significantly independent factor for the patients with LUAD (HR = 1.449, 95% CI = 1.312 - 1.602, P < 0.001). Conclusion The nine hypoxia-related lncRNAs and their signature might be molecular biomarkers and therapeutic targets for the patients with LUAD.
Collapse
Affiliation(s)
- Jiaojiao Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | | | - Hui Zhang
- Department of Medical Imaging, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Qiang Song
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Bo Guo
- Institute of Genetics and Developmental Biology, Translational Medicine Institute, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Jin Shang
- Department of Medical Imaging, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
7
|
Quarleri J, Delpino MV. The interplay of aging, adipose tissue, and COVID-19: a potent alliance with implications for health. GeroScience 2024; 46:2915-2932. [PMID: 38191833 PMCID: PMC11009220 DOI: 10.1007/s11357-023-01058-z] [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: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024] Open
Abstract
Obesity has emerged as a significant public health challenge. With the ongoing increase in life expectancy, the prevalence of obesity is steadily growing, particularly among older age demographics. The extension of life expectancy frequently results in additional years of vulnerability to chronic health issues associated with obesity in the elderly.The concept of SARS-CoV-2 directly infecting adipose tissue stems from the fact that both adipocytes and stromal vascular fraction cells express ACE2, the primary receptor facilitating SARS-CoV-2 entry. It is noteworthy that adipose tissue demonstrates ACE2 expression levels similar to those found in the lungs within the same individual. Additionally, ACE2 expression in the adipose tissue of obese individuals surpasses that in non-obese counterparts. Viral attachment to ACE2 has the potential to disturb the equilibrium of renin-angiotensin system homeostasis, leading to an exacerbated inflammatory response.Consequently, adipose tissue has been investigated as a potential site for active SARS-CoV-2 infection, suggesting its plausible role in virus persistence and contribution to both acute and long-term consequences associated with COVID-19.This review is dedicated to presenting current evidence concerning the presence of SARS-CoV-2 in the adipose tissue of elderly individuals infected with the virus. Both obesity and aging are circumstances that contribute to severe health challenges, heightening the risk of disease and mortality. We will particularly focus on examining the mechanisms implicated in the long-term consequences, with the intention of providing insights into potential strategies for mitigating the aftermath of the disease.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Paraguay 2155, Piso 11, C1121ABG, Ciudad Autónoma de Buenos Aires, Argentina.
| | - M Victoria Delpino
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Paraguay 2155, Piso 11, C1121ABG, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
8
|
Zhang D, Wang Y, Zhou Z, Wang L, Liu C, Jiang Y. Role of miRNA-regulated type H vessel formation in osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1394785. [PMID: 38883597 PMCID: PMC11176424 DOI: 10.3389/fendo.2024.1394785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Osteoporosis (OP) is a chronic systemic bone metabolism disease characterized by decreased bone mass, microarchitectural deterioration, and fragility fractures. With the demographic change caused by long lifespans and population aging, OP is a growing health problem. The role of miRNA in the pathogenesis of OP has also attracted widespread attention from scholars in recent years. Type H vessels are unique microvessels of the bone and have become a new focus in the pathogenesis of OP because they play an essential role in osteogenesis-angiogenesis coupling. Previous studies found some miRNAs regulate type H vessel formation through the regulatory factors, including platelet-derived growth factor-BB (PDGF-BB), hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF), and so on. These findings help us gain a more in-depth understanding of the relationship among miRNAs, type H vessels, and OP to find a new perspective on treating OP. In the present mini-review, we will introduce the role of type H vessels in the pathogenesis of OP and the regulation of miRNAs on type H vessel formation by affecting regulatory factors to provide some valuable insights for future studies of OP treatment.
Collapse
Affiliation(s)
- Dailiang Zhang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yongjing Wang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Zunzhen Zhou
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Limei Wang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Chongzhi Liu
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yuan Jiang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Vajdi M, Karimi A, Hassanizadeh S, Farhangi MA, Bagherniya M, Askari G, Roufogalis BD, Davies NM, Sahebkar A. Effect of polyphenols against complications of COVID-19: current evidence and potential efficacy. Pharmacol Rep 2024; 76:307-327. [PMID: 38498260 DOI: 10.1007/s43440-024-00585-6] [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: 09/23/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024]
Abstract
The COVID-19 pandemic that started in 2019 and resulted in significant morbidity and mortality continues to be a significant global health challenge, characterized by inflammation, oxidative stress, and immune system dysfunction.. Developing therapies for preventing or treating COVID-19 remains an important goal for pharmacology and drug development research. Polyphenols are effective against various viral infections and can be extracted and isolated from plants without losing their therapeutic potential. Researchers have developed methods for separating and isolating polyphenols from complex matrices. Polyphenols are effective in treating common viral infections, including COVID-19, and can also boost immunity. Polyphenolic-based antiviral medications can mitigate SARS-CoV-2 enzymes vital to virus replication and infection. Individual polyphenolic triterpenoids, flavonoids, anthraquinonoids, and tannins may also inhibit the SARS-CoV-2 protease. Polyphenol pharmacophore structures identified to date can explain their action and lead to the design of novel anti-COVID-19 compounds. Polyphenol-containing mixtures offer the advantages of a well-recognized safety profile with few known severe side effects. However, studies to date are limited, and further animal studies and randomized controlled trials are needed in future studies. The purpose of this study was to review and present the latest findings on the therapeutic impact of plant-derived polyphenols on COVID-19 infection and its complications. Exploring alternative approaches to traditional therapies could aid in developing novel drugs and remedies against coronavirus infection.
Collapse
Affiliation(s)
- Mahdi Vajdi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Karimi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shirin Hassanizadeh
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Abbasalizad Farhangi
- Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Bagherniya
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Basil D Roufogalis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Zhang Z, Zhang N, Lu X, Zhou M, Yan X, Gu W, Yang J, Zhang Q, Zhang C, Gong Y, Jia M, Zhang X, Ning P, Liu M, Li X, Shi X, Liu W, Gao GF, Ning G, Wang J, Bi Y. Anti-infection effects of heparin on SARS-CoV-2 in a diabetic mouse model. Zool Res 2023; 44:1003-1014. [PMID: 37759335 PMCID: PMC10802103 DOI: 10.24272/j.issn.2095-8137.2023.108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can result in more severe syndromes and poorer outcomes in patients with diabetes and obesity. However, the precise mechanisms responsible for the combined impact of corona virus disease 2019 (COVID-19) and diabetes have not yet been elucidated, and effective treatment options for SARS-CoV-2-infected diabetic patients remain limited. To investigate the disease pathogenesis, K18-hACE2 transgenic (hACE2 Tg) mice with a leptin receptor deficiency (hACE2-Lepr -/-) or high-fat diet (hACE2-HFD) background were generated. The two mouse models were intranasally infected with a 5×10 5 median tissue culture infectious dose (TCID 50) of SARS-CoV-2, with serum and lung tissue samples collected at 3 days post-infection. The hACE2-Lepr -/- mice were then administered a combination of low-molecular-weight heparin (LMWH) (1 mg/kg or 5 mg/kg) and insulin via subcutaneous injection prior to intranasal infection with 1×10 4 TCID 50 of SARS-CoV-2. Daily drug administration continued until the euthanasia of the mice. Analyses of viral RNA loads, histopathological changes in lung tissue, and inflammation factors were conducted. Results demonstrated similar SARS-CoV-2 susceptibility in hACE2 Tg mice under both lean (chow diet) and obese (HFD) conditions. However, compared to the hACE2-Lepr +/+ mice, hACE2-Lepr -/- mice exhibited more severe lung injury, enhanced expression of inflammatory cytokines and hypoxia-inducible factor-1α, and increased apoptosis. Moreover, combined LMWH and insulin treatment effectively reduced disease progression and severity, attenuated lung pathological changes, and mitigated inflammatory responses. In conclusion, pre-existing diabetes can lead to more severe lung damage upon SARS-CoV-2 infection, and LMWH may be a valuable therapeutic approach for managing COVID-19 patients with diabetes.
Collapse
Affiliation(s)
- Zhongyun Zhang
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Ning Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Xuancheng Lu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxiang Yan
- Department of Cardiology, Institute of Cardiovascular Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiqiong Gu
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jingru Yang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qin Zhang
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Cheng Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Yuhuan Gong
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Mingjun Jia
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Ning
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
| | - Mei Liu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Xiaoyan Li
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Xiaomeng Shi
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guang Ning
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China. E-mail:
| | - Yuhai Bi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), CAS-TWAS Center of Excellence for Emerging Infectious Diseases (CEEID), Chinese Academy of Sciences, Beijing 100101, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- University of Chinese Academy of Sciences, Beijing 100049, China. E-mail:
| |
Collapse
|
11
|
Leyderman M, Wilmore JR, Shope T, Cooney RN, Urao N. Impact of intestinal microenvironments in obesity and bariatric surgery on shaping macrophages. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00033. [PMID: 38037591 PMCID: PMC10683977 DOI: 10.1097/in9.0000000000000033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Obesity is associated with alterations in tissue composition, systemic cellular metabolism, and low-grade chronic inflammation. Macrophages are heterogenous innate immune cells ubiquitously localized throughout the body and are key components of tissue homeostasis, inflammation, wound healing, and various disease states. Macrophages are highly plastic and can switch their phenotypic polarization and change function in response to their local environments. Here, we discuss how obesity alters the intestinal microenvironment and potential key factors that can influence intestinal macrophages as well as macrophages in other organs, including adipose tissue and hematopoietic organs. As bariatric surgery can induce metabolic adaptation systemically, we discuss the potential mechanisms through which bariatric surgery reshapes macrophages in obesity.
Collapse
Affiliation(s)
- Michael Leyderman
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Joel R. Wilmore
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Timothy Shope
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Robert N. Cooney
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Norifumi Urao
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
12
|
Jin Y, Ren W, Liu J, Tang X, Shi X, Pan D, Hou L, Yang L. Identification and validation of potential hypoxia-related genes associated with coronary artery disease. Front Physiol 2023; 14:1181510. [PMID: 37637145 PMCID: PMC10447898 DOI: 10.3389/fphys.2023.1181510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction: Coronary artery disease (CAD) is one of the most life-threatening cardiovascular emergencies with high mortality and morbidity. Increasing evidence has demonstrated that the degree of hypoxia is closely associated with the development and survival outcomes of CAD patients. However, the role of hypoxia in CAD has not been elucidated. Methods: Based on the GSE113079 microarray dataset and the hypoxia-associated gene collection, differential analysis, machine learning, and validation of the screened hub genes were carried out. Results: In this study, 54 differentially expressed hypoxia-related genes (DE-HRGs), and then 4 hub signature genes (ADM, PPFIA4, FAM162A, and TPBG) were identified based on microarray datasets GSE113079 which including of 93 CAD patients and 48 healthy controls and hypoxia-related gene set. Then, 4 hub genes were also validated in other three CAD related microarray datasets. Through GO and KEGG pathway enrichment analyses, we found three upregulated hub genes (ADM, PPFIA4, TPBG) were strongly correlated with differentially expressed metabolic genes and all the 4 hub genes were mainly enriched in many immune-related biological processes and pathways in CAD. Additionally, 10 immune cell types were found significantly different between the CAD and control groups, especially CD8 T cells, which were apparently essential in cardiovascular disease by immune cell infiltration analysis. Furthermore, we compared the expression of 4 hub genes in 15 cell subtypes in CAD coronary lesions and found that ADM, FAM162A and TPBG were all expressed at higher levels in endothelial cells by single-cell sequencing analysis. Discussion: The study identified four hypoxia genes associated with coronary heart disease. The findings provide more insights into the hypoxia landscape and, potentially, the therapeutic targets of CAD.
Collapse
Affiliation(s)
- Yuqing Jin
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Weiyan Ren
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Jiayi Liu
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xuejiao Tang
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Xinrui Shi
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Dongchen Pan
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Lianguo Hou
- Biochemistry Research Laboratory, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Lei Yang
- Department of Epidemiology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
13
|
Howlader DR, Das S, Lu T, Mandal RS, Hu G, Varisco DJ, Dietz ZK, Ratnakaram SSK, Ernst RK, Picking WD, Picking WL. A protein subunit vaccine elicits a balanced immune response that protects against Pseudomonas pulmonary infection. NPJ Vaccines 2023; 8:37. [PMID: 36918600 PMCID: PMC10012293 DOI: 10.1038/s41541-023-00618-w] [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: 09/06/2022] [Accepted: 02/02/2023] [Indexed: 03/15/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa (Pa) causes severe nosocomial infections, especially in immunocompromised individuals and the elderly. Increasing drug resistance, the absence of a licensed vaccine and increased hospitalizations due to SARS-CoV-2 have made Pa a major healthcare risk. To address this, we formulated a candidate subunit vaccine against Pa (L-PaF), by fusing the type III secretion system tip and translocator proteins with LTA1 in an oil-in-water emulsion (ME). This was mixed with the TLR4 agonist (BECC438b). Lung mRNA sequencing showed that the formulation activates genes from multiple immunological pathways eliciting a protective Th1-Th17 response following IN immunization. Following infection, however, the immunized mice showed an adaptive response while the PBS-vaccinated mice experienced rapid onset of an inflammatory response. The latter displayed a hypoxic lung environment with high bacterial burden. Finally, the importance of IL-17 and immunoglobulins were demonstrated using knockout mice. These findings suggest a need for a balanced humoral and cellular response to prevent the onset of Pa infection and that our formulation could elicit such a response.
Collapse
Affiliation(s)
- Debaki R Howlader
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Sayan Das
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, 21201, USA
| | - Ti Lu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Rahul Shubhra Mandal
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gang Hu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA
| | - David J Varisco
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, 21201, USA
| | - Zackary K Dietz
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | | | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, 21201, USA
| | - William D Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Wendy L Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA.
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
14
|
Sun K, Li X, Scherer PE. Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives. Compr Physiol 2023; 13:4387-4407. [PMID: 36715281 PMCID: PMC9957663 DOI: 10.1002/cphy.c220020] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer. © 2023 American Physiological Society. Compr Physiol 13:4387-4407, 2023.
Collapse
Affiliation(s)
- Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
15
|
Al‐Kuraishy HM, Al‐Gareeb AI, Mohammed AA, Alexiou A, Papadakis M, Batiha GE. The potential link between Covid-19 and multiple myeloma: A new saga. Immun Inflamm Dis 2022; 10:e701. [PMID: 36444620 PMCID: PMC9673426 DOI: 10.1002/iid3.701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Covid-19 is considered a primary respiratory disease-causing viral pneumonia and, in severe cases, leads to acute lung injury and acute respiratory distress syndrome (ARDS). In addition, though, extra-pulmonary manifestations of Covid-19 have been shown. Furthermore, severe acute respiratory distress syndrome coronavirus type 2 (SARS-CoV-2) infection may coexist with several malignancies, including multiple myeloma (MM). METHODS This critical literature review aimed to find the potential association between SARS-CoV-2 infection and MM in Covid-19 patients with underlying MM. Narrative literature and databases search revealed that ARDS is developed in both MM and Covid-19 due to hypercalcemia and proteasome dysfunction. RESULTS Notably, the expression of angiogenic factors and glutamine deficiency could link Covid-19 severity and MM in the pathogenesis of cardiovascular complications. MM and Covid-19 share thrombosis as a typical complication; unlike thrombosis in Covid-19, which reflects disease severity, thrombosis does not reflect disease severity in MM. In both conditions, thromboprophylaxis is essential to prevent pulmonary thrombosis and other thromboembolic disorders. Moreover, Covid-19 may exacerbate the development of acute kidney injury and neurological complications in MM patients. CONCLUSION These findings highlighted that MM patients might be a risk group for Covid-19 severity due to underlying immunosuppression and most of those patients need specific management in the Covid-19 era.
Collapse
Affiliation(s)
- Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali A Mohammed
- The Chest Clinic, Barts Health NHS TrustWhipps Cross University HospitalLondonUK
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
16
|
Yucel K, Fuat Gurbuz A. Hypoxia-inducible factor-1α and ischemia-modified albumin levels in intensive care COVID-19 Patients. Horm Mol Biol Clin Investig 2022; 43:415-420. [PMID: 35851469 DOI: 10.1515/hmbci-2022-0024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/07/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES In this study, it was aimed to evaluate the hypoxia-inducible factor-1α (HIF-1α) and ischemia-modified albumin (IMA) levels of patients diagnosed with COVID-19 in the intensive care unit (ICU) and healthy controls. To our knowledge, this is the first study investigate HIF-1α and IMA levels in COVID-19 patients in ICUs and comparing them with a healthy control group. For this reason, our study is original and will contribute to the literature. METHODS A total of 70 intensive care patients diagnosed with COVID-19, and 72 healthy controls were included in the study. RESULTS When we compared the patient and healthy control group; there were no statistically significant differences between the groups in terms of age and gender (p>0.05). No exitus was observed in the patient group. We found weak correlation between HIF-1α and IMA (r: 0.320). However, there were statistically significant differences in HIF-1α and IMA levels in the patient group. The receiver operating characteristic (ROC) curve demonstrated an area under curve (AUC) value of 0.651 for HIF-1α and 0.937 for IMA. CONCLUSIONS The HIF-1α and IMA levels were significantly higher among COVID-19 patients in ICU compared with healthy controls. HIF-1α and IMA levels can be used as reliable markers for the prognosis of COVID-19.
Collapse
Affiliation(s)
- Kamile Yucel
- Department of Medical Biochemistry, KTO Karatay University, Faculty of Medicine, Konya, Turkey
| | - Ali Fuat Gurbuz
- Department of Internal Medicine, Health Sciences University, Van Training and Research Hospital, Van, Turkey
| |
Collapse
|
17
|
Mortezaee K, Majidpoor J. Cellular immune states in SARS-CoV-2-induced disease. Front Immunol 2022; 13:1016304. [PMID: 36505442 PMCID: PMC9726761 DOI: 10.3389/fimmu.2022.1016304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
The general immune state plays important roles against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Cells of the immune system are encountering rapid changes during the acute phase of SARS-CoV-2-induced disease. Reduced fraction of functional CD8+ T cells, disrupted cross-talking between CD8+ T cells with dendritic cells (DCs), and impaired immunological T-cell memory, along with the higher presence of hyperactive neutrophils, high expansion of myeloid-derived suppressor cells (MDSCs) and non-classical monocytes, and attenuated cytotoxic capacity of natural killer (NK) cells, are all indicative of low efficient immunity against viral surge within the body. Immune state and responses from pro- or anti-inflammatory cells of the immune system to SARS-CoV-2 are discussed in this review. We also suggest some strategies to enhance the power of immune system against SARS-CoV-2-induced disease.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran,*Correspondence: Keywan Mortezaee, ;
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
18
|
Yang C, Zhou Y, Liu H, Xu P. The Role of Inflammation in Cognitive Impairment of Obstructive Sleep Apnea Syndrome. Brain Sci 2022; 12:brainsci12101303. [PMID: 36291237 PMCID: PMC9599901 DOI: 10.3390/brainsci12101303] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) has become a major worldwide public health concern, given its global prevalence. It has clear links with multiple comorbidities and mortality. Cognitive impairment is one related comorbidity causing great pressure on individuals and society. The clinical manifestations of cognitive impairment in OSAS include decline in attention/vigilance, verbal–visual memory loss, visuospatial/structural ability impairment, and executive dysfunction. It has been proven that chronic intermittent hypoxia (CIH) may be a main cause of cognitive impairment in OSAS. Inflammation plays important roles in CIH-induced cognitive dysfunction. Furthermore, the nuclear factor kappa B and hypoxia-inducible factor 1 alpha pathways play significant roles in this inflammatory mechanism. Continuous positive airway pressure is an effective therapy for OSAS; however, its effect on cognitive impairment is suboptimal. Therefore, in this review, we address the role inflammation plays in the development of neuro-impairment in OSAS and the association between OSAS and cognitive impairment to provide an overview of its pathophysiology. We believe that furthering the understanding of the inflammatory mechanisms involved in OSAS-associated cognitive impairment could lead to the development of appropriate and effective therapy.
Collapse
|
19
|
Brahmi F, Vejux A, Ghzaiel I, Ksila M, Zarrouk A, Ghrairi T, Essadek S, Mandard S, Leoni V, Poli G, Vervandier-Fasseur D, Kharoubi O, El Midaoui A, Atanasov AG, Meziane S, Latruffe N, Nasser B, Bouhaouala-Zahar B, Masmoudi-Kouki O, Madani K, Boulekbache-Makhlouf L, Lizard G. Role of Diet and Nutrients in SARS-CoV-2 Infection: Incidence on Oxidative Stress, Inflammatory Status and Viral Production. Nutrients 2022; 14:2194. [PMID: 35683996 PMCID: PMC9182601 DOI: 10.3390/nu14112194] [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: 04/03/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
Coronavirus illness (COVID-19) is an infectious pathology generated by intense severe respiratory syndrome coronavirus 2 (SARS-CoV-2). This infectious disease has emerged in 2019. The COVID-19-associated pandemic has considerably affected the way of life and the economy in the world. It is consequently crucial to find solutions allowing remedying or alleviating the effects of this infectious disease. Natural products have been in perpetual application from immemorial time given that they are attested to be efficient towards several illnesses without major side effects. Various studies have shown that plant extracts or purified molecules have a promising inhibiting impact towards coronavirus. In addition, it is substantial to understand the characteristics, susceptibility and impact of diet on patients infected with COVID-19. In this review, we recapitulate the influence of extracts or pure molecules from medicinal plants on COVID-19. We approach the possibilities of plant treatment/co-treatment and feeding applied to COVID-19. We also show coronavirus susceptibility and complications associated with nutrient deficiencies and then discuss the major food groups efficient on COVID-19 pathogenesis. Then, we covered emerging technologies using plant-based SARS-CoV-2 vaccine. We conclude by giving nutrient and plants curative therapy recommendations which are of potential interest in the COVID-19 infection and could pave the way for pharmacological treatments or co-treatments of COVID-19.
Collapse
Affiliation(s)
- Fatiha Brahmi
- Laboratory Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria; (K.M.); (L.B.-M.)
| | - Anne Vejux
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
| | - Imen Ghzaiel
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
- Lab-NAFS ‘Nutrition-Functional Food & Vascular Health’, Faculty of Medicine, LR12ES05, University Monastir, Monastir 5000, Tunisia;
| | - Mohamed Ksila
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia; (T.G.); (O.M.-K.)
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition-Functional Food & Vascular Health’, Faculty of Medicine, LR12ES05, University Monastir, Monastir 5000, Tunisia;
- Laboratory of Biochemistry, Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
| | - Taoufik Ghrairi
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia; (T.G.); (O.M.-K.)
| | - Soukena Essadek
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
- Laboratory Neuroscience and Biochemistry, University of Hassan 1st, Settat 26000, Morocco;
| | - Stéphane Mandard
- Lipness Team and LipSTIC LabEx, UFR Sciences de Santé, INSERM/University of Bourgogne Franche-Comté LNC UMR1231, 21000 Dijon, France;
| | - Valerio Leoni
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale Brianza ASST-Brianza, Desio Hospital, Via Mazzini 1, 20833 Desio, Italy;
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Turin, 10043 Orbassano (Turin), Italy;
| | - Dominique Vervandier-Fasseur
- Team OCS, Institute of Molecular Chemistry of University of Burgundy (ICMUB UMR CNRS 6302), University of Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Omar Kharoubi
- Laboratory of Experimental Biotoxicology, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences, University Oran 1 ABB, Oran 31000, Algeria;
| | - Adil El Midaoui
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada;
- Faculty of Sciences and Techniques, Moulay Ismail University of Meknes, Errachidia 52000, Morocco
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland;
| | - Smail Meziane
- Institut Européen des Antioxydants, 1b Rue Victor de Lespinats, 54230 Neuves-Maison, France;
| | - Norbert Latruffe
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
| | - Boubker Nasser
- Laboratory Neuroscience and Biochemistry, University of Hassan 1st, Settat 26000, Morocco;
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Biomolecules, Venoms and Theranostic Applications, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia;
| | - Olfa Masmoudi-Kouki
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia; (T.G.); (O.M.-K.)
| | - Khodir Madani
- Laboratory Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria; (K.M.); (L.B.-M.)
- Centre de Recherche en Technologie des Industries Agroalimentaires, Route de Targua Ouzemour, Bejaia 06000, Algeria
| | - Lila Boulekbache-Makhlouf
- Laboratory Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria; (K.M.); (L.B.-M.)
| | - Gérard Lizard
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
| |
Collapse
|
20
|
AbdelMassih A, Gaber H, El Shershaby M, Hanafy M, Omar Y, Husseiny R, AlShehry N, Ismail HA, Kamel A, Hozaien R, Khaled G, Amer M, Turki A, Fawzy H, Puligheddu S, Khaled D, Thabet NN, Abdelaziz MS, Barakat M, Sharaf S, Mohamed A, Mohsen D, El Feky A, Adly H, Ibrahim E, Mahmoud R, Reda M, Riad F, Vasile C, Shohdi MA, Hesham N, El-Husseiny N, Ragy R, Fouda R. Learned lesson from COVID-19: can routine immunizations be the first line of defense against the next pandemic? EGYPTIAN PEDIATRIC ASSOCIATION GAZETTE 2022. [PMCID: PMC8978766 DOI: 10.1186/s43054-022-00105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Single-cell sequencing studies on the lung microenvironment have revealed that the outcome of COVID-19 depends largely on the immune system response rather than the viral load. A robust innate immune response and a regulated adaptive immunity can prevent the worst outcomes such as hospitalization and the need for mechanical ventilation.
Main body
Intriguingly, several vaccines pertaining to the routine vaccination schedule, not only BCG, can skew the immune response towards the aforementioned beneficial effects.
Short conclusion
This means that routine immunization not only can help in the current pandemic but can also offer a rapid rescue in the subsequent epidemics or pandemics until a vaccine is developed.
Collapse
|
21
|
Chiner-Vives E, Cordovilla-Pérez R, de la Rosa-Carrillo D, García-Clemente M, Izquierdo-Alonso JL, Otero-Candelera R, Pérez-de Llano L, Sellares-Torres J, de Granda-Orive JI. Short and Long-Term Impact of COVID-19 Infection on Previous Respiratory Diseases. Arch Bronconeumol 2022; 58 Suppl 1:39-50. [PMID: 35501222 PMCID: PMC9012323 DOI: 10.1016/j.arbres.2022.03.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
On March 11, 2020, the World Health Organization declared Coronavirus Disease 2019 (COVID-19) a pandemic. Till now, it affected 452.4 million (Spain, 11.18 million) persons all over the world with a total of 6.04 million of deaths (Spain, 100,992). It is observed that 75% of hospitalized COVID-19 patients have at least one COVID-19 associated comorbidity. It was shown that people with underlying chronic illnesses are more likely to get it and grow seriously ill. Individuals with COVID-19 who have a past medical history of cardiovascular disorder, cancer, obesity, chronic lung disease, diabetes, or neurological disease had the worst prognosis and are more likely to develop acute respiratory distress syndrome or pneumonia. COVID-19 can affect the respiratory system in a variety of ways and across a spectrum of levels of disease severity, depending on a person's immune system, age and comorbidities. Symptoms can range from mild, such as cough, shortness of breath and fever, to critical disease, including respiratory failure, shock and multi-organ system failure. So, COVID-19 infection can cause overall worsening of these previous respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), interstitial lung disease, etc. This review aims to provide information on the impact of the COVID-19 disease on pre-existing lung comorbidities.
Collapse
Affiliation(s)
- Eusebi Chiner-Vives
- Multidisciplinary Sleep Unit, Respiratory Department, Sant Joan University Hospital, Sant Joan d'Alacant, Alicante, Spain
| | - Rosa Cordovilla-Pérez
- Respiratory Department, Salamanca University Hospital, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | | | - Marta García-Clemente
- Lung Management Area, HUCA, Institute for Health Research of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
| | - José Luis Izquierdo-Alonso
- Department of Medicine and Medical Specialties, University of Alcalá, Madrid, Spain; Respiratory Medicine, University Hospital of Guadalajara, Guadalajara, Spain
| | | | - Luis Pérez-de Llano
- Respiratory Department, Lucus Augusti University Hospital, EOXI Lugo, Monforte, CERVO, Lugo, Spain
| | - Jacobo Sellares-Torres
- Interstitial Lung Diseases Working Group, Respiratory Department, Clinic-University Hospital-IDIBAPS, Barcelona, Spain
| | | |
Collapse
|
22
|
Mortezaee K, Majidpoor J. CD8 + T Cells in SARS-CoV-2 Induced Disease and Cancer-Clinical Perspectives. Front Immunol 2022; 13:864298. [PMID: 35432340 PMCID: PMC9010719 DOI: 10.3389/fimmu.2022.864298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Dysregulated innate and adaptive immunity is a sign of SARS-CoV-2-induced disease and cancer. CD8+ T cells are important cells of the immune system. The cells belong to the adaptive immunity and take a front-line defense against viral infections and cancer. Extreme CD8+ T-cell activities in the lung of patients with a SARS-CoV-2-induced disease and within the tumor microenvironment (TME) will change their functionality into exhausted state and undergo apoptosis. Such diminished immunity will put cancer cases at a high-risk group for SARS-CoV-2-induced disease, rendering viral sepsis and a more severe condition which will finally cause a higher rate of mortality. Recovering responses from CD8+ T cells is a purpose of vaccination against SARS-CoV-2. The aim of this review is to discuss the CD8+ T cellular state in SARS-CoV-2-induced disease and in cancer and to present some strategies for recovering the functionality of these critical cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
23
|
Shafqat A, Shafqat S, Salameh SA, Kashir J, Alkattan K, Yaqinuddin A. Mechanistic Insights Into the Immune Pathophysiology of COVID-19; An In-Depth Review. Front Immunol 2022; 13:835104. [PMID: 35401519 PMCID: PMC8989408 DOI: 10.3389/fimmu.2022.835104] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), which causes coronavirus-19 (COVID-19), has caused significant morbidity and mortality globally. In addition to the respiratory manifestations seen in severe cases, multi-organ pathologies also occur, making management a much-debated issue. In addition, the emergence of new variants can potentially render vaccines with a relatively limited utility. Many investigators have attempted to elucidate the precise pathophysiological mechanisms causing COVID-19 respiratory and systemic disease. Spillover of lung-derived cytokines causing a cytokine storm is considered the cause of systemic disease. However, recent studies have provided contradictory evidence, whereby the extent of cytokine storm is insufficient to cause severe illness. These issues are highly relevant, as management approaches considering COVID-19 a classic form of acute respiratory distress syndrome with a cytokine storm could translate to unfounded clinical decisions, detrimental to patient trajectory. Additionally, the precise immune cell signatures that characterize disease of varying severity remain contentious. We provide an up-to-date review on the immune dysregulation caused by COVID-19 and highlight pertinent discussions in the scientific community. The response from the scientific community has been unprecedented regarding the development of highly effective vaccines and cutting-edge research on novel therapies. We hope that this review furthers the conversations held by scientists and informs the aims of future research projects, which will potentially further our understanding of COVID-19 and its immune pathogenesis.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Center of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | |
Collapse
|
24
|
Potential of Polyphenols to Restore SIRT1 and NAD+ Metabolism in Renal Disease. Nutrients 2022; 14:nu14030653. [PMID: 35277012 PMCID: PMC8837945 DOI: 10.3390/nu14030653] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/18/2022] [Accepted: 01/29/2022] [Indexed: 11/17/2022] Open
Abstract
SIRT1 is an NAD+-dependent class III histone deacetylase that is abundantly expressed in the kidney, where it modulates gene expression, apoptosis, energy homeostasis, autophagy, acute stress responses, and mitochondrial biogenesis. Alterations in SIRT1 activity and NAD+ metabolism are frequently observed in acute and chronic kidney diseases of diverse origins, including obesity and diabetes. Nevertheless, in vitro and in vivo studies and clinical trials with humans show that the SIRT1-activating compounds derived from natural sources, such as polyphenols found in fruits, vegetables, and plants, including resveratrol, quercetin, and isoflavones, can prevent disease and be part of treatments for a wide variety of diseases. Here, we summarize the roles of SIRT1 and NAD+ metabolism in renal pathophysiology and provide an overview of polyphenols that have the potential to restore SIRT1 and NAD+ metabolism in renal diseases.
Collapse
|
25
|
Luu R, Valdebenito S, Scemes E, Cibelli A, Spray DC, Rovegno M, Tichauer J, Cottignies-Calamarte A, Rosenberg A, Capron C, Belouzard S, Dubuisson J, Annane D, de la Grandmaison GL, Cramer-Bordé E, Bomsel M, Eugenin E. Pannexin-1 channel opening is critical for COVID-19 pathogenesis. iScience 2021; 24:103478. [PMID: 34841222 PMCID: PMC8603863 DOI: 10.1016/j.isci.2021.103478] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/30/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly rampaged worldwide, causing a pandemic of coronavirus disease (COVID -19), but the biology of SARS-CoV-2 remains under investigation. We demonstrate that both SARS-CoV-2 spike protein and human coronavirus 229E (hCoV-229E) or its purified S protein, one of the main viruses responsible for the common cold, induce the transient opening of Pannexin-1 (Panx-1) channels in human lung epithelial cells. However, the Panx-1 channel opening induced by SARS-CoV-2 is greater and more prolonged than hCoV-229E/S protein, resulting in an enhanced ATP, PGE2, and IL-1β release. Analysis of lung lavages and tissues indicate that Panx-1 mRNA expression is associated with increased ATP, PGE2, and IL-1β levels. Panx-1 channel opening induced by SARS-CoV-2 spike protein is angiotensin-converting enzyme 2 (ACE-2), endocytosis, and furin dependent. Overall, we demonstrated that Panx-1 channel is a critical contributor to SARS-CoV-2 infection and should be considered as an alternative therapy.
Collapse
Affiliation(s)
- Ross Luu
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX 77555, USA
| | - Silvana Valdebenito
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX 77555, USA
| | - Eliana Scemes
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - Antonio Cibelli
- Dominick P. Purpura Department of Neuroscience & Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - David C Spray
- Dominick P. Purpura Department of Neuroscience & Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Tichauer
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrea Cottignies-Calamarte
- Hôpital Cochin, Service de Virologie, Hôpital Cochin (AP-HP), Paris, France.,Service d'Hématologie Hôpital Ambroise Paré (AP-HP), Boulogne-Billancourt, France
| | - Arielle Rosenberg
- Hôpital Cochin, Service de Virologie, Hôpital Cochin (AP-HP), Paris, France.,Service d'Hématologie Hôpital Ambroise Paré (AP-HP), Boulogne-Billancourt, France.,Virologie Moléculaire et Cellulaire des Coronavirus, Centre d'infection et d'immunité de Lille, Institut Pasteur de Lille, Université de Lille, CNRS, Inserm, CHRU, 59000 Lille, France
| | - Calude Capron
- Service des Maladies Infectieuses, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | | | - Jean Dubuisson
- Intensive Care Unit, Raymond Poincaré Hospital (AP-HP), Paris, France
| | - Djillali Annane
- Simone Veil School of Medicine, Université of Versailles, Versailles, France.,University Paris Saclay, Garches, France
| | - Geoffroy Lorin de la Grandmaison
- Department of Forensic Medicine and Pathology, Versailles Saint-Quentin Université, AP-HP, Raymond Poincaré Hospital, Garches, France
| | | | - Morgane Bomsel
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France.,INSERM U1016, Paris, France
| | - Eliseo Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Research Building 17, 105 11th Street, Galveston, TX 77555, USA
| |
Collapse
|
26
|
Hasankhani A, Bahrami A, Sheybani N, Aria B, Hemati B, Fatehi F, Ghaem Maghami Farahani H, Javanmard G, Rezaee M, Kastelic JP, Barkema HW. Differential Co-Expression Network Analysis Reveals Key Hub-High Traffic Genes as Potential Therapeutic Targets for COVID-19 Pandemic. Front Immunol 2021; 12:789317. [PMID: 34975885 PMCID: PMC8714803 DOI: 10.3389/fimmu.2021.789317] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background The recent emergence of COVID-19, rapid worldwide spread, and incomplete knowledge of molecular mechanisms underlying SARS-CoV-2 infection have limited development of therapeutic strategies. Our objective was to systematically investigate molecular regulatory mechanisms of COVID-19, using a combination of high throughput RNA-sequencing-based transcriptomics and systems biology approaches. Methods RNA-Seq data from peripheral blood mononuclear cells (PBMCs) of healthy persons, mild and severe 17 COVID-19 patients were analyzed to generate a gene expression matrix. Weighted gene co-expression network analysis (WGCNA) was used to identify co-expression modules in healthy samples as a reference set. For differential co-expression network analysis, module preservation and module-trait relationships approaches were used to identify key modules. Then, protein-protein interaction (PPI) networks, based on co-expressed hub genes, were constructed to identify hub genes/TFs with the highest information transfer (hub-high traffic genes) within candidate modules. Results Based on differential co-expression network analysis, connectivity patterns and network density, 72% (15 of 21) of modules identified in healthy samples were altered by SARS-CoV-2 infection. Therefore, SARS-CoV-2 caused systemic perturbations in host biological gene networks. In functional enrichment analysis, among 15 non-preserved modules and two significant highly-correlated modules (identified by MTRs), 9 modules were directly related to the host immune response and COVID-19 immunopathogenesis. Intriguingly, systemic investigation of SARS-CoV-2 infection identified signaling pathways and key genes/proteins associated with COVID-19's main hallmarks, e.g., cytokine storm, respiratory distress syndrome (ARDS), acute lung injury (ALI), lymphopenia, coagulation disorders, thrombosis, and pregnancy complications, as well as comorbidities associated with COVID-19, e.g., asthma, diabetic complications, cardiovascular diseases (CVDs), liver disorders and acute kidney injury (AKI). Topological analysis with betweenness centrality (BC) identified 290 hub-high traffic genes, central in both co-expression and PPI networks. We also identified several transcriptional regulatory factors, including NFKB1, HIF1A, AHR, and TP53, with important immunoregulatory roles in SARS-CoV-2 infection. Moreover, several hub-high traffic genes, including IL6, IL1B, IL10, TNF, SOCS1, SOCS3, ICAM1, PTEN, RHOA, GDI2, SUMO1, CASP1, IRAK3, HSPA5, ADRB2, PRF1, GZMB, OASL, CCL5, HSP90AA1, HSPD1, IFNG, MAPK1, RAB5A, and TNFRSF1A had the highest rates of information transfer in 9 candidate modules and central roles in COVID-19 immunopathogenesis. Conclusion This study provides comprehensive information on molecular mechanisms of SARS-CoV-2-host interactions and identifies several hub-high traffic genes as promising therapeutic targets for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Nuclear Agriculture Research School, Nuclear Science and Technology Research Institute, Karaj, Iran
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Behzad Aria
- Department of Physical Education and Sports Science, School of Psychology and Educational Sciences, Yazd University, Yazd, Iran
| | - Behzad Hemati
- Biotechnology Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Farhang Fatehi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | | | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mahsa Rezaee
- Department of Medical Mycology, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
27
|
Yao J, Du Y, Liu J, Gareev I, Yang G, Kang X, Wang X, Beylerli O, Chen X. Hypoxia related long non-coding RNAs in ischemic stroke. Noncoding RNA Res 2021; 6:153-158. [PMID: 34703955 PMCID: PMC8511691 DOI: 10.1016/j.ncrna.2021.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/02/2021] [Accepted: 10/02/2021] [Indexed: 12/22/2022] Open
Abstract
With high rates of mortality and disability, stroke has caused huge social burden, and 85% of which is ischemic stroke. In recent years, it is a progressive discovery of long non-coding RNA (lncRNA) playing an important regulatory role throughout ischemic stroke. Hypoxia, generated from reduction or interruption of cerebral blood flow, leads to changes in lncRNA expression, which then influence disease progression. Therefore, we reviewed studies on expression of hypoxia-related lncRNAs and relevant molecular mechanism in ischemic stroke. Considering that hypoxia-inducible factor (HIF) is a crucial regulator in hypoxic progress, we mainly focus on the HIF-related lncRNA which regulates the expression of HIF or is regulated by HIF, further reveal their pathogenesis and adaption after brain ischemia and hypoxia, so as to find effective biomarker and therapeutic targets.
Collapse
Affiliation(s)
- Jiawei Yao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yiming Du
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Junsi Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Ilgiz Gareev
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Guang Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Xiaohui Kang
- Department of Pharmacy, Rizhao People's Hospital, Rizhao, 276826, Shandong Province, China
| | - Xiaoxiong Wang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Ozal Beylerli
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| |
Collapse
|
28
|
Shirvaliloo M. The blood-gas barrier in COVID-19: an overview of the effects of SARS-CoV-2 infection on the alveolar epithelial and endothelial cells of the lung. Tissue Barriers 2021; 9:1937013. [PMID: 34232823 PMCID: PMC8794501 DOI: 10.1080/21688370.2021.1937013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 02/08/2023] Open
Abstract
Blood-gas barrier (BGB) or alveolar-capillary barrier is the primary tissue barrier affected by coronavirus disease 2019 (COVID-19). Comprising alveolar epithelial cells (AECs), endothelial cells (ECs) and the extracellular matrix (ECM) in between, the BGB is damaged following the action of multiple pro-inflammatory cytokines during acute inflammation. The infection of AECs and ECs with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen behind COVID-19, triggers an inflammatory response at the BGB, inducing the release of interleukin 1 (IL-1), IL-6, tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-β), high mobility group box 1 (HMGB1), matrix metalloproteinases (MMPs), intercellular adhesion molecule-1 (ICAM-1) and platelet activating factor (PAF). The end result is the disassembly of adherens junctions (AJs) and tight junctions (TJs) in both AECs and ECs, AEC hyperplasia, EC pyroptosis, ECM remodeling and deposition of fibrin clots in the alveolar capillaries, leading to disintegration and thickening of the BGB, and ultimately, hypoxia. This commentary seeks to provide a brief account of how the BGB might become affected in COVID-19.
Collapse
Affiliation(s)
- Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Das A, Patra M, Dhangadamajhi G. Association of rs11549465 (C1772T) variant of hypoxia-inducible factor-1α with Covid-19 susceptibility. A population-based epidemiological study. Hum Cell 2021; 34:1937-1940. [PMID: 34426956 PMCID: PMC8382105 DOI: 10.1007/s13577-021-00601-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Anamika Das
- Agricultural and Food Engineering Department, Indian Institute of Technology, Kharagpur, 721302, West Bengal, India
| | - Maheswari Patra
- Department of Biotechnology, Maharaja Sriram Chandra Bhanjadeo University, Baripada, 757003, Odisha, India
| | - Gunanidhi Dhangadamajhi
- Department of Biotechnology, Maharaja Sriram Chandra Bhanjadeo University, Baripada, 757003, Odisha, India.
| |
Collapse
|
30
|
Ninomiya T, Otsubo K, Hoshino T, Shimokawa M, Nakazawa M, Sato Y, Mikumo H, Kawakami S, Mizusaki S, Mori Y, Arimura H, Tsuchiya-Kawano Y, Inoue K, Uchida Y, Nakanishi Y. Risk factors for disease progression in Japanese patients with COVID-19 with no or mild symptoms on admission. BMC Infect Dis 2021; 21:850. [PMID: 34419004 PMCID: PMC8379599 DOI: 10.1186/s12879-021-06574-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Although the risk factors for coronavirus disease 2019 (COVID-19) mortality have been identified, there is limited information about the risk factors for disease progression after hospitalization among Japanese patients with COVID-19 exhibiting no or mild symptoms. METHODS All 302 consecutive patients who were admitted to our institutions and diagnosed with COVID-19 between March and December 2020 were retrospectively assessed. Ultimately, 210 adult patients exhibiting no or mild symptoms on admission were included in the analysis. They were categorized into the stable (no oxygen needed) and worsened (oxygen needed) groups, and their characteristics and laboratory data were compared. RESULTS Among 210 patients, 49 progressed to a severe disease stage, whereas 161 did not. The mean patient age was 52.14 years, and 126 (60.0%) patients were male. The mean body mass index (BMI) was 23.0 kg/m2, and 71 patients were overweight (BMI ≥ 25 kg/m2). Multivariate logistic analysis showed that old age, overweight, diabetes mellitus (DM), and high serum ferritin levels were independent risk factors for disease progression. CONCLUSIONS Clinicians should closely observe patients with COVID-19, especially those with risk factors such as old age, overweight, DM, and high serum ferritin levels, regardless of whether they have no or mild symptoms.
Collapse
Affiliation(s)
- Toshifumi Ninomiya
- Department of Respiratory Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan.,Department of General Internal Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Kohei Otsubo
- Department of Respiratory Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan.
| | - Teppei Hoshino
- Department of Internal Medicine, Kitakyushu City Yahata Hospital, 2-6-2, Ogura, Yahatahigashi-ku, Kitakyushu, Fukuoka, 805-8534, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube-shi, Yamaguchi, 755-8505, Japan
| | - Megumi Nakazawa
- Department of General Internal Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Yoriko Sato
- Department of General Internal Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Hironori Mikumo
- Department of General Internal Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Satoru Kawakami
- Department of General Internal Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan.,Department of Respiratory Medicine, JCHO Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-ku, Kitakyushu, Fukuoka, 806-8501, Japan
| | - Shun Mizusaki
- Department of Respiratory Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan.,Department of General Internal Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Yusuke Mori
- Department of Internal Medicine, Kitakyushu City Yahata Hospital, 2-6-2, Ogura, Yahatahigashi-ku, Kitakyushu, Fukuoka, 805-8534, Japan
| | - Hidenobu Arimura
- Department of Respiratory Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Yuko Tsuchiya-Kawano
- Department of Respiratory Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Koji Inoue
- Department of Respiratory Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Yujiro Uchida
- Department of General Internal Medicine, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, Fukuoka, 802-0077, Japan
| | - Yoichi Nakanishi
- Kitakyushu City Hospital Organization, 1-35 Furusenbamachi, Kokurakita-ku, Kitakyushu, 802-0082, Japan
| |
Collapse
|
31
|
Gusev E, Sarapultsev A, Hu D, Chereshnev V. Problems of Pathogenesis and Pathogenetic Therapy of COVID-19 from the Perspective of the General Theory of Pathological Systems (General Pathological Processes). Int J Mol Sci 2021; 22:7582. [PMID: 34299201 PMCID: PMC8304657 DOI: 10.3390/ijms22147582] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 01/18/2023] Open
Abstract
The COVID-19 pandemic examines not only the state of actual health care but also the state of fundamental medicine in various countries. Pro-inflammatory processes extend far beyond the classical concepts of inflammation. They manifest themselves in a variety of ways, beginning with extreme physiology, then allostasis at low-grade inflammation, and finally the shockogenic phenomenon of "inflammatory systemic microcirculation". The pathogenetic core of critical situations, including COVID-19, is this phenomenon. Microcirculatory abnormalities, on the other hand, lie at the heart of a specific type of general pathological process known as systemic inflammation (SI). Systemic inflammatory response, cytokine release, cytokine storm, and thrombo-inflammatory syndrome are all terms that refer to different aspects of SI. As a result, the metabolic syndrome model does not adequately reflect the pathophysiology of persistent low-grade systemic inflammation (ChSLGI). Diseases associated with ChSLGI, on the other hand, are risk factors for a severe COVID-19 course. The review examines the role of hypoxia, metabolic dysfunction, scavenger receptors, and pattern-recognition receptors, as well as the processes of the hemophagocytic syndrome, in the systemic alteration and development of SI in COVID-19.
Collapse
Affiliation(s)
- Evgenii Gusev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (E.G.); (V.C.)
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (E.G.); (V.C.)
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 200092, China;
| | - Valeriy Chereshnev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (E.G.); (V.C.)
| |
Collapse
|
32
|
Zhu Z, Zheng Z, Liu J. Comparison of COVID-19 and Lung Cancer via Reactive Oxygen Species Signaling. Front Oncol 2021; 11:708263. [PMID: 34277453 PMCID: PMC8283805 DOI: 10.3389/fonc.2021.708263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022] Open
Abstract
COVID-19 and lung cancer are two severe pulmonary diseases that cause millions of deaths globally each year. Understanding the dysregulated signaling pathways between them can benefit treating the related patients. Recent studies suggest the critical role of reactive oxygen species (ROS) in both diseases, indicating an interplay between them. Here we reviewed references showing that ROS and ROS-associated signaling pathways, specifically via NRF2, HIF-1, and Nf-κB pathways, may bridge mutual impact between COVID-19 and lung cancer. As expected, typical ROS-associated inflammation pathways (HIF-1 and Nf-κB) are activated in both diseases. The activation of both pathways in immune cells leads to an overloading immune response and exacerbates inflammation in COVID-19. In lung cancer, HIF-1 activation facilitates immune escape, while Nf-κB activation in T cells suppresses tumor growth. However, the altered NRF2 pathway show opposite trends between them, NRF2 pathways exert immunosuppressive effects in both diseases, as it represses the immune response in COVID-19 patients while facilitates the immune escape of tumor cells. Furthermore, we summarized the therapeutic targets (e.g., phytochemicals) on these ROS pathways. In sum, our review focus on the understanding of ROS Signaling in COVID-19 and lung cancer, showing that modulating ROS signaling pathways may alleviate the potentially mutual impacts between COVID-19 and lung cancer patients.
Collapse
Affiliation(s)
- Zilan Zhu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Ziyi Zheng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| |
Collapse
|
33
|
Chirumbolo S, Valdenassi L, Simonetti V, Bertossi D, Ricevuti G, Franzini M, Pandolfi S. Insights on the mechanisms of action of ozone in the medical therapy against COVID-19. Int Immunopharmacol 2021; 96:107777. [PMID: 34020394 PMCID: PMC8112288 DOI: 10.1016/j.intimp.2021.107777] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/28/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
An increasing amount of reports in the literature is showing that medical ozone (O3) is used, with encouraging results, in treating COVID-19 patients, optimizing pain and symptoms relief, respiratory parameters, inflammatory and coagulation markers and the overall health status, so reducing significantly how much time patients underwent hospitalization and intensive care. To date, aside from mechanisms taking into account the ability of O3 to activate a rapid oxidative stress response, by up-regulating antioxidant and scavenging enzymes, no sound hypothesis was addressed to attempt a synopsis of how O3 should act on COVID-19. The knowledge on how O3 works on inflammation and thrombosis mechanisms is of the utmost importance to make physicians endowed with new guns against SARS-CoV2 pandemic. This review tries to address this issue, so to expand the debate in the scientific community.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Luigi Valdenassi
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy
| | - Vincenzo Simonetti
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy
| | - Dario Bertossi
- Department of Surgery, Dentistry, Paediatrics and Gynaecology Unit of Maxillo-Facial Surgery University of Verona, Verona, Italy
| | | | - Marianno Franzini
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy
| | - Sergio Pandolfi
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy; Villa Mafalda Clinics via Monte delle Gioie, Rome, Italy
| |
Collapse
|
34
|
Berdina ON, Madaeva IM, Rychkova LV. [Novel coronavirus disease (COVID-19) and obstructive sleep apnea: age aspects of comorbidity]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:110-115. [PMID: 34078869 DOI: 10.17116/jnevro2021121402110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In connection with the spread of the novel coronavirus infection (COVID-19) pandemic and the increase in the development of severe acute respiratory syndrome, works are published around the world that determined the risk factors for complications and poor outcomes in this disease. Among the main comorbidities in COVID-19, scientists distinguish hypertension, diabetes, obesity, etc. Recently, more and more physicians and researchers are concerned about the high frequency of severe and critical complications of COVID-19 in patients with obstructive sleep apnea (OSA). In this review, we present some data on COVID-19 and OSA in the age aspect, show the general pathophysiological pathways leading to increased systemic inflammation and adverse consequences in the comorbid course of these diseases. Special attention is paid to such studies in the pediatric population, but only a few works of foreign scientists were found that did not reveal the essence of the problem under discussion, which requires further research in this area.
Collapse
Affiliation(s)
- O N Berdina
- Scientific Centre for Family Health and Human Reproduction problems, Irkutsk, Russia
| | - I M Madaeva
- Scientific Centre for Family Health and Human Reproduction problems, Irkutsk, Russia
| | - L V Rychkova
- Scientific Centre for Family Health and Human Reproduction problems, Irkutsk, Russia
| |
Collapse
|
35
|
Vaishali K, Gatty A, Srivastav P, Amin RR. Coping strategies for obese individuals with obstructive sleep apnea during COVID-19 pandemic: A narrative review. ACTA ACUST UNITED AC 2021; 22:100324. [PMID: 33589881 PMCID: PMC7874942 DOI: 10.1016/j.obmed.2021.100324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/24/2021] [Accepted: 02/02/2021] [Indexed: 01/08/2023]
Abstract
Background Patients infected with SARS-CoV-2- having pre-existing non-communicable diseases (NCDs)- are at a higher risk of complications. Obesity is one of the proven risk factors causing NCDs and can influence outcomes of COVID-19 patients. It is closely related to obstructive sleep apnea (OSA). The increased risk of COVID-19 and reduced access to treatment of non-COVID conditions during the pandemic may increase the stress in obese patients with OSA. This situation makes it necessary for them to cope with their condition by themselves. This review aimed at the effect of this pandemic on these patients and coping strategies for them. Methods Databases like PubMed and Scopus were searched using a combination of key words. Full-text articles meeting the inclusion criteria were selected. Results The search yielded eight studies, discussing about the potential interactions between the COVID-19, obesity and OSA, the impact of COVID-19 on them, and management of these patients. Conclusions Increased prevalence of COVID-19 was found among obese patients with OSA. The fear of COVID-19 and shift of health care workers to manage COVID-19 patients has affected their regular visits to the hospital. However, there is lack of coping strategies for them, which should soon be established for these patients.
Collapse
Affiliation(s)
- K Vaishali
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Aishwarya Gatty
- College of Physiotherapy, Srinivas University, Mangalore, India
| | - Prateek Srivastav
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Revati Ravi Amin
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education (MAHE), Manipal, India
| |
Collapse
|