1
|
Klimova EM, Lavinska OV, Drozdova LA. The role of DAMP cytotoxic fractions in the immune markers' disruption in patients with urgent surgical pathology and against the background of post-COVID-19 syndrome. Immunol Lett 2025:107033. [PMID: 40409597 DOI: 10.1016/j.imlet.2025.107033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND As a result of the SARS-CoV-2 pandemic, various population groups were formed that had acute and asymptomatic COVID-19. A survey in these groups revealed with equal frequency an asthenic symptom complex, the so-called post-COVID-19 syndrome (PCS). The frequency of urgent surgical pathology against the background of PCS and structural and functional disorders of various organs was increased. The aim - to study the dynamics of immunoresistance factors changes in patients with urgent surgical pathology that developed against the background of PCS and to identify pathogenic markers of the severe course and the risk of mortality. MATERIALS AND METHODS To examine patients with PCS and urgent cardiovascular (n = 103) and abdominal (n = 106) pathology we used the following methods: fluorescence microscopy, confocal microscopy, flow cytometry, spectrophotometry, ELISA. RESULTS We revealed a temporal dependence of immune dysfunction in patients with a comorbid course of urgent surgical pathology and PCS. The nature of the DAMP (damage-associated molecular patterns) cytotoxic fractions ratio was associated with certain changes in innate and adaptive immunity factors, severity of the condition and risk of mortality. At the first stage (2020-2021), patients with PCS has disorders of the humoral and cellular components of innate immunity against the background of an increase in the oligopeptide and peptide DAMP fractions. At the second stage (2022-2024) of PCS development, changes in innate as well as adaptive immunity were observed against the background of an increase in the cytotoxic oligonucleotide DAMP fraction (mortality was 17.3%). CONCLUSIONS The identified markers of impaired immunoresistance in cardiovascular and abdominal urgent pathology can be used to select targeted therapy tactics.
Collapse
Affiliation(s)
- Elena M Klimova
- Department of Molecular Biology and Biotechnology, V.N. Karazin Kharkiv National University, 4 Pl. Svobody, 61022 Kharkiv, Ukraine; Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine.
| | - Olena V Lavinska
- Department of Molecular Biology and Biotechnology, V.N. Karazin Kharkiv National University, 4 Pl. Svobody, 61022 Kharkiv, Ukraine; Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine
| | - Larisa A Drozdova
- Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine
| |
Collapse
|
2
|
Gasmi M, Hejazi M, Muscella A, Marsigliante S, Sharma A. Aging-associated changes in immunological parameters: Implications for COVID-19 immune response in the elderly. Physiol Rep 2025; 13:e70364. [PMID: 40405557 PMCID: PMC12098970 DOI: 10.14814/phy2.70364] [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: 01/29/2025] [Revised: 04/07/2025] [Accepted: 04/26/2025] [Indexed: 05/24/2025] Open
Abstract
Aging has a profound impact on the immune system, leading to a gradual decline in its function and increased systemic inflammation, collectively known as immunosenescence and inflammaging. These changes make older adults more susceptible to infections, including COVID-19, and contribute to worse clinical outcomes, such as higher morbidity and mortality rates. This review explores immunological changes associated with aging, including impaired innate immune responses, reduced T- and B-cell function, and altered cytokine profiles. A comprehensive literature search identified relevant studies on the topic, and inclusion criteria focused on studies addressing age-related immune changes and their impact on responses to COVID-19. The findings underscore the need for targeted healthcare strategies to mitigate the negative effects of aging on immunity and improve immune resilience, and ultimately clinical outcomes and quality of life for this vulnerable population.
Collapse
Affiliation(s)
- Maha Gasmi
- Higher Institute of Sport and Physical Education of Ksar SaidTunisTunisia
| | - Mahdi Hejazi
- Department of Nutrition, School of Public HealthIran University of Medical SciencesTehranIran
| | - Antonella Muscella
- Department of Biological and Environmental Science and Technologies (DiSTeBA)University of SalentoLecceItaly
| | - Santo Marsigliante
- Department of Biological and Environmental Science and Technologies (DiSTeBA)University of SalentoLecceItaly
| | - Aastha Sharma
- Department of Basic and Applied Science, School of Engineering and ScienceUniversity‐GD Goenka University GurugramGurugramIndia
| |
Collapse
|
3
|
Wang Y, Fu Q, Sha S, Yoon S. Interferon inhibitors increase rAAV production in HEK293 cells. J Biotechnol 2025; 399:9-18. [PMID: 39824361 DOI: 10.1016/j.jbiotec.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 12/17/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Recombinant adeno-associated viruses (rAAVs) comprise a promising viral vector for therapeutic gene delivery to treat disease. However, the current manufacturing capability of rAAVs must be improved to meet commercial demand. Previously published omics studies indicate that rAAV production through transient transfection triggers antiviral responses and endoplasmic reticulum stress responses in the host cell. Both responses negatively regulate viral production. We demonstrate that the modulation of the antiviral immune response (by blocking interferon signaling pathways) can effectively lower the production of interferon and enhance viral genome production. The use of interferon inhibitors before transfection can significantly increase rAAV production in HEK293 cells, with up to a 2-fold increase in productivity and up to a 6-fold increase in specific productivity. Compared to the untreated groups, the addition of these small molecules generally reduced viable cell density but increased vector productivity. The positive candidates were BX795 (a TBK inhibitor), TPCA-1 (an IKK2 inhibitor), Cyt387 (a JAK1 inhibitor), and ruxolitinib (another JAK1 inhibitor). These candidates were identified using deep well screening, and reproducible titer improvement was achieved in a 30 mL shake flask scale. Additionally, genome titer improvement is feasible and scalable in two different media, but the extent of improvement may vary.
Collapse
Affiliation(s)
- Yongdan Wang
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | - Qiang Fu
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | - Sha Sha
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, United States.
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, United States.
| |
Collapse
|
4
|
Lee MJ, Litchford ML, Vendrame E, Vergara R, Ranganath T, Fish CS, Chebet D, Langat A, Mburu C, Neary J, Benki S, Wamalwa D, John-Stewart G, Lehman DA, Blish CA. Distinct immune profiles in children living with HIV based on timing and duration of suppressive antiretroviral treatment. Virology 2025; 602:110318. [PMID: 39612623 PMCID: PMC11645197 DOI: 10.1016/j.virol.2024.110318] [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: 09/17/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Timely initiation of antiretroviral therapy (ART) remains a major challenge in the effort to treat children living with HIV ("CLH") and little is known regarding the dynamics of immune normalization following ART in CLH with varying times to and durations of ART. Here, we leveraged two cohorts of virally-suppressed CLH from Nairobi, Kenya to examine differences in the peripheral immune systems between two cohorts of age-matched children (to control for immune changes with age): one group which initiated ART during early HIV infection and had been on ART for 5-6 years at evaluation (early, long-term treated; "ELT" cohort), and one group which initiated ART later and had been on ART for approximately 9 months at evaluation (delayed, short-term treated; "DST" cohort). We profiled PBMC and purified NK cells from these two cohorts by mass cytometry time-of-flight (CyTOF). Although both groups of CLH had undetectable viral RNA load at evaluation, there were marked differences in both immune composition and immune phenotype between the ELT cohort and the DST cohort. DST donors had reduced CD4 T cell percentages, decreased naive to effector memory T cell ratios, and markedly higher expression of stress-induced markers. Conversely, ELT donors had higher naive to effector memory T cell ratios, low expression of stress-induced markers, and increased expression of markers associated with an effective antiviral response and resolution of inflammation. Collectively, our results demonstrate key differences in the immune systems of virally-suppressed CLH with different ages at ART initiation and durations of treatment and provide further rationale for emphasizing early onset of ART.
Collapse
Affiliation(s)
- Madeline J Lee
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Morgan L Litchford
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elena Vendrame
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosemary Vergara
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Thanmayi Ranganath
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Carolyn S Fish
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daisy Chebet
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Agnes Langat
- Division of Global HIV & TB., Center for Global Health, U.S Centers for Disease Control and Prevention, USA
| | - Caren Mburu
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Jillian Neary
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Sarah Benki
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Dalton Wamalwa
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | | | - Dara A Lehman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
5
|
Wang D, Wang K, Liu Q, Liu M, Zhang G, Feng K, Wang K, Ding X, Zhu H, Yang S, Liu Y, Li T, Gong P, Wang M, Wang PG, Jin H, Zhao W, Yu F. A Novel Drug Candidate for Sepsis Targeting Heparanase by Inhibiting Cytokine Storm. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403337. [PMID: 38810101 PMCID: PMC11304236 DOI: 10.1002/advs.202403337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 05/31/2024]
Abstract
Sepsis is an infection-triggered, rapidly progressive systemic inflammatory syndrome with a high mortality rate. Currently, there are no promising therapeutic strategies for managing this disease in the clinic. Heparanase plays a crucial role in the pathology of sepsis, and its inhibition can significantly relieve related symptoms. Here, a novel heparanase inhibitor CV122 is rationally designed and synthesized, and its therapeutic potential for sepsis with Lipopolysaccharide (LPS) and Cecal Ligation and Puncture (CLP)-induced sepsis mouse models are evaluated. It is found that CV122 potently inhibits heparanase activity in vitro, protects cell surface glycocalyx structure, and reduces the expression of adhesion molecules. In vivo, CV122 significantly reduces the systemic levels of proinflammatory cytokines, prevents organ damage, improves vitality, and efficiently protects mice from sepsis-induced death. Mechanistically, CV122 inhibits the activity of heparanase, reduces its expression in the lungs, and protects glycocalyx structure of lung tissue. It is also found that CV122 provides effective protection from organ damage and death caused by Crimean-Congo hemorrhagic fever virus (CCHFV) infection. These results suggest that CV122 is a potential drug candidate for sepsis therapy targeting heparanase by inhibiting cytokine storm.
Collapse
Affiliation(s)
- Danyang Wang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Kaixuan Wang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Qiutong Liu
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Mingyang Liu
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Guoqiang Zhang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Ke Feng
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Kun Wang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Xianwei Ding
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Haomiao Zhu
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Song Yang
- School of Health and Life SciencesQingdao Central HospitalUniversity of Health and Rehabilitation SciencesQingdao266113China
| | - Yonghui Liu
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Tiehai Li
- Carbohydrate‐Based Drug Research CenterShanghai Institute of Materia MedicalChinese Academy of SciencesShanghai201203China
| | - Peng Gong
- State Key Laboratory of VirologyWuhan Institute of VirologyChinese Academy of SciencesWuhan430071China
| | - Manli Wang
- State Key Laboratory of VirologyWuhan Institute of VirologyChinese Academy of SciencesWuhan430071China
| | - Peng George Wang
- School of MedicineSouthern University of Science and TechnologyShenzhen518000China
| | - Hongzhen Jin
- School of Health and Life SciencesQingdao Central HospitalUniversity of Health and Rehabilitation SciencesQingdao266113China
| | - Wei Zhao
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyKey Laboratory of Molecular Drug Research and KLMDASR of TianjinNankai UniversityTongyan Road, Haihe Education ParkTianjin300350China
| | - Fan Yu
- School of Health and Life SciencesQingdao Central HospitalUniversity of Health and Rehabilitation SciencesQingdao266113China
| |
Collapse
|
6
|
Lu J, Chen Y, Zhou K, Ling Y, Qin Q, Lu W, Qin L, Mou C, Zhang J, Zheng X, Qin K. Immune characteristics of kidney transplant recipients with acute respiratory distress syndrome induced by COVID-19 at single-cell resolution. Respir Res 2024; 25:34. [PMID: 38238762 PMCID: PMC10795319 DOI: 10.1186/s12931-024-02682-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND COVID-19-induced acute respiratory distress syndrome (ARDS) can result in tissue damage and multiple organ dysfunction, especially in kidney transplant recipients (KTRs) receiving immunosuppressive drugs. Presently, single-cell research on COVID-19-induced ARDS is considerably advanced, yet knowledge about ARDS in KTRs is still constrained. METHODS Single-cell RNA sequencing (scRNA-seq) analysis was performed to construct a comprehensive single-cell immune landscape of the peripheral blood mononuclear cells (PBMCs) of eight patients with COVID-19-induced ARDS, five KTRs with COVID-19-induced ARDS, and five healthy individuals. Subsequently, we conducted a comprehensive bioinformatics analysis, including cell clustering, enrichment analysis, trajectory analysis, gene regulatory network analysis, and cell-cell interaction analysis, to investigate the heterogeneity of the immune microenvironment in KTRs with ARDS. RESULT Our study revealed that KTRs exhibit significant heterogeneity with COVID-19-induced ARDS compared with those of other individuals, with significant reductions in T cells, as well as an abnormal proliferation of B cells and monocytes. In the context of dual influences from immunosuppression and viral infection, KTRs exhibited more specific plasma cells, along with significant enrichment of dysfunctional GZMB and XAF1 double-positive effector T cells and IFI27-positive monocytes. Additionally, robust communication existed among T cells and monocytes in cytokine signaling. These effects impede the process of immune reconstitution in KTR patients. CONCLUSION Our findings suggest that KTRs with COVID-19-induced ARDS show elevated antibody levels, impaired T cell differentiation, and dysregulation of innate immunity. In summary, this study provides a theoretical foundation for a comprehensive understanding of COVID-19-induced ARDS in KTRs.
Collapse
Affiliation(s)
- Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
- Guangxi Health Commission Key Laboratory of Emergency and Critical Medicine, Nanning, 530007, China.
| | - Yin Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Yicong Ling
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Qianqian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Weisheng Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Lian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Chenglin Mou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Jianfeng Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
- Guangxi Health Commission Key Laboratory of Emergency and Critical Medicine, Nanning, 530007, China
| | - Xiaowen Zheng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
- Guangxi Health Commission Key Laboratory of Emergency and Critical Medicine, Nanning, 530007, China.
| | - Ke Qin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
- Department of Anesthesiology, Guilin People's Hospital, Guilin, 541002, China.
| |
Collapse
|
7
|
Jin J, Wang X, Li Y, Yang X, Wang H, Han X, Sun J, Ma Z, Duan J, Zhang G, Huang T, Zhang T, Wu H, Zhang X, Su B. Weak SARS-CoV-2-specific responses of TIGIT-expressing CD8 + T cells in people living with HIV after a third dose of a SARS-CoV-2 inactivated vaccine. Chin Med J (Engl) 2023; 136:2938-2947. [PMID: 37963586 PMCID: PMC10752475 DOI: 10.1097/cm9.0000000000002926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibition motif domains (TIGIT), an inhibitory receptor expressed on T cells, plays a dysfunctional role in antiviral infection and antitumor activity. However, it is unknown whether TIGIT expression on T cells influences the immunological effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) inactivated vaccines. METHODS Forty-five people living with HIV (PLWH) on antiretroviral therapy (ART) for more than two years and 31 healthy controls (HCs), all received a third dose of a SARS-CoV-2 inactivated vaccine, were enrolled in this study. The amounts, activation, proportion of cell subsets, and magnitude of the SARS-CoV-2-specific immune response of TIGIT + CD4 + and TIGIT + CD8 + T cells were investigated before the third dose but 6 months after the second vaccine dose (0W), 4 weeks (4W) and 12 weeks (12W) after the third dose. RESULTS Compared to that in HCs, the frequency of TIGIT + CD8 + T cells in the peripheral blood of PLWH increased at 12W after the third dose of the inactivated vaccine, and the immune activation of TIGIT + CD8 + T cells also increased. A decrease in the ratio of both T naïve (T N ) and central memory (T CM ) cells among TIGIT + CD8 + T cells and an increase in the ratio of the effector memory (T EM ) subpopulation were observed at 12W in PLWH. Interestingly, particularly at 12W, a higher proportion of TIGIT + CD8 + T cells expressing CD137 and CD69 simultaneously was observed in HCs than in PLWH based on the activation-induced marker assay. Compared with 0W, SARS-CoV-2-specific TIGIT + CD8 + T-cell responses in PLWH were not enhanced at 12W but were enhanced in HCs. Additionally, at all time points, the SARS-CoV-2-specific responses of TIGIT + CD8 + T cells in PLWH were significantly weaker than those of TIGIT - CD8 + T cells. However, in HCs, the difference in the SARS-CoV-2-specific responses induced between TIGIT + CD8 + T cells and TIGIT - CD8 + T cells was insignificant at 4W and 12W, except at 0W. CONCLUSIONS TIGIT expression on CD8 + T cells may hinder the T-cell immune response to a booster dose of an inactivated SARS-CoV-2 vaccine, suggesting weakened resistance to SARS-CoV-2 infection, especially in PLWH. Furthermore, TIGIT may be used as a potential target to increase the production of SARS-CoV-2-specific CD8 + T cells, thereby enhancing the effectiveness of vaccination.
Collapse
Affiliation(s)
- Junyan Jin
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Yongzheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100871, China
| | - Xiaodong Yang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Hu Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xiaoxu Han
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Jin Sun
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Zhenglai Ma
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Junyi Duan
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Guanghui Zhang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tao Huang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
8
|
Sadik J, Sharma M, Deshmukh CV, Maqhbool SB, Thekiya AH, Kamble PP. Assessment of Genotoxic Biomarker in Tongue and Buccal Mucosal Epithelial Cells of COVID-19 Patients: An Observational Study. Cureus 2023; 15:e48706. [PMID: 38094540 PMCID: PMC10716604 DOI: 10.7759/cureus.48706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2023] [Indexed: 04/27/2025] Open
Abstract
INTRODUCTION Angiotensin-converting enzyme 2 (ACE2) is the main host cell receptor for coronavirus disease 2019 (COVID-19) and is highly expressed in the tongue and buccal mucosa. Therefore, the present study was conducted to investigate genotoxic changes in epithelial cells of the buccal and tongue mucosa following COVID-19 infection. MATERIALS AND METHODS This study included 40 patients aged 25-40 years, divided into two groups: Group 1 (control group) included 20 healthy individuals with no prior history of COVID-19 infection subdivided into Group 1a (buccal mucosa), and Group 1b (tongue mucosa); Group 2 (case group) included 20 patients with a history of mild to moderate COVID-19 infection subdivided into Group 2a (buccal mucosa) and Group 2b (tongue mucosa). Genotoxic biomarkers, such as the number of micronuclei, pyknosis, karyolysis, and karyorrhexis, were assessed in epithelial cells from the buccal mucosa and the ventral surface of the tongue. Analysis of variance was used for intragroup comparisons, followed by post-hoc analysis using Tukey's test. RESULTS The mean age of the patients was 27.4±6.52 years. Statistically significant differences were observed between cases and controls in the number of micronuclei, pyknosis, karyolysis, and karyorrhexis in the epithelial cells of the buccal and tongue mucosa (p = 0.05). CONCLUSION SARS-CoV-2 has pronounced genotoxic effects on the epithelium of the ventral surface of the tongue in comparison to the buccal mucosa Therefore, patients with COVID-19 should be monitored regularly to develop future carcinomas, particularly those with habits of smoking, alcohol consumption, and tobacco usage.
Collapse
Affiliation(s)
- Jafer Sadik
- Department of Dentistry, Government Hospital, Periyur, IND
| | - Manish Sharma
- Department of Oral Pathology, Jawahar Medical Foundation (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| | - Chetan V Deshmukh
- Department of Public Health Dentistry, Nair Hospital Dental College, Mumbai, IND
| | | | - Altaf H Thekiya
- Department of Orthodontics and Dentofacial Orthopedics, Diamond Dental Care, Nanded, IND
| | - Priyanka P Kamble
- Department of Oral Pathology, Jawahar Medical Foundation (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| |
Collapse
|
9
|
Ali MO, Alva B, Nagaral S, Patil R, Khan MU, Tiwari DA. Association Between Candida albicans and COVID-19 in Complete Denture Wearers: An Observational Study. Cureus 2023; 15:e47777. [PMID: 38022199 PMCID: PMC10676466 DOI: 10.7759/cureus.47777] [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] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The phenomenon of coronavirus disease 2019 (COVID-19)-related candidiasis is gaining increased attention and acknowledgment as an integral component of the severe consequences of COVID-19. The aim of the present study was to assess the association between Candida albicans and COVID-19 in complete denture wearers. Materials and methods An observational study was conducted on 45 complete denture wearers, who were divided into three groups as follows: Group 1, 15 subjects with mild to moderate COVID-19 infection; Group 2, 15 subjects with severe COVID-19 infection; and Group 3, 15 subjects without COVID-19 infection. Mean colony forming units (CFU) were observed on agar plates containing Sabouraud dextrose in the salivary samples of the participants. Analysis of variance, followed by post-hoc analysis by Tukey's test, was used to compare CFU between the groups. Pearson's correlation coefficient was used to study the correlation between variables. Results The highest average colony-forming units of Candida albicans were observed in Group 2, followed by Group 1, compared to the control group, and a significant (p<0.001) difference was found. A weak positive correlation was found between the age of the patients and the duration of denture usage, as well as between age and the counts of Candida albicans in Groups 1 and 3. This correlation was more pronounced in Group 3. A strong positive correlation was observed in all groups between the Candida albicans count and the duration of denture usage by the patients. Conclusion The association between Candida albicans and denture wear was compounded by the presence of COVID-19. Consequently, the timely identification of Candida albicans infection in patients with COVID-19 is important to establish more efficacious approaches for antifungal treatment and prophylactic interventions.
Collapse
Affiliation(s)
- Mohd Osman Ali
- Department of Dentistry, Deccan College of Medical Sciences, Hyderabad, IND
| | - Babashankar Alva
- Department of Dentistry, Faculty of Dentistry, Ramaiah University of Applied Sciences, Bangaluru, IND
| | - Suresh Nagaral
- Department of Prosthodontics, JMF's ACPM Dental College, Dhule, IND
| | - Rohit Patil
- Department of Prosthodontics, JMF's ACPM Dental College, Dhule, IND
| | | | - Durgesh A Tiwari
- Department of Conservative Dentistry and Endodontics, Yogita Dental College and Hospital, Khed, IND
| |
Collapse
|
10
|
Xin X, Yao W, Zhang Z, Yang X, Li S, Zhu Y, Zhang C, Zhang L, Huang H, Dong T, Dong H, Feng L, Wang S. Immune and cytokine alterations and RNA-sequencing analysis in gestational tissues from pregnant women after recovery from COVID-19. BMC Infect Dis 2023; 23:620. [PMID: 37735363 PMCID: PMC10512579 DOI: 10.1186/s12879-023-08607-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND COVID-19 is a global pandemic. Understanding the immune responses in pregnant women recovering from COVID-19 may suggest new therapeutic approaches. METHODS We performed a cross-sectional study between March 1, 2020, and September 1, 2020. Participants were assigned into the convalescent COVID-19 group if they had a previous COVID-19 infection during pregnancy or the healthy control group. RNA-Seq was performed on human umbilical cord mesenchymal stem cells (hUMSCs) and human amniotic mesenchymal stem cells (hAMSCs). Immunohistochemical staining, cytokine testing, lymphocyte subset analysis, RNA-Seq, and functional analyses were performed on the placental and umbilical cord blood (UCB) and compared between the two groups. RESULTS A total of 40 pregnant women were enrolled, with 13 in the convalescent group and 27 in the control group. There were 1024, 46, and 32 differentially expressed genes (DEGs) identified in the placental tissue, hUMSCs, and hAMSCs between the convalescent and control groups, respectively. Enrichment analysis showed those DEGs were associated with immune homeostasis, antiviral activity, cell proliferation, and tissue repair. Levels of IL-6, TNF-α, total lymphocyte counts, B lymphocytes, Tregs percentages, and IFN-γ expressing CD4+ and CD8+ T cells were statistically different between two groups (p ≤ 0.05). ACE2 and TMPRSS2 expressed on the placenta were not different between the two groups (p > 0.05). CONCLUSION Multiple changes in immune responses occurred in the placental tissue, hUMSCs, and hAMSCs after maternal recovery from COVID-19, which might imply their protective roles against COVID-19 infection.
Collapse
Affiliation(s)
- Xing Xin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China
| | - Weiqi Yao
- Wuhan Optics Valley Vcanbiopharma Co. Ltd, Wuhan, 430000, Hubei, P.R. China
- VCANBIO Cell & Gene Engineering Corp., Ltd, Tianjin, 300384, P.R. China
- Department of Biology and medicine, Hubei University of Technology, Wuhan, 430068, Hubei, P.R. China
- Wuhan Optics Valley Vcanbio Cell & Gene Technology Co., Ltd, Wuhan, 430000, Hubei, P.R. China
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, 430000, Hubei, P.R. China
| | - Zijing Zhang
- Department of Medical office, Wuchang Shouyi College Hospital, Wuhan, 430064, Hubei, P.R. China
| | - Xin Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China
| | - Shufang Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China
| | - Ying Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China
| | - Cong Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China
| | - Long Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China
| | - Hailong Huang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430077, Hubei, P.R. China
| | - Tengyun Dong
- Wuhan Optics Valley Vcanbiopharma Co. Ltd, Wuhan, 430000, Hubei, P.R. China
- Wuhan Optics Valley Vcanbio Cell & Gene Technology Co., Ltd, Wuhan, 430000, Hubei, P.R. China
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, 430000, Hubei, P.R. China
| | - Haibo Dong
- Wuhan Optics Valley Vcanbio Cell & Gene Technology Co., Ltd, Wuhan, 430000, Hubei, P.R. China
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, 430000, Hubei, P.R. China
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China.
| | - Shaoshuai Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430032, Hubei, P.R. China.
| |
Collapse
|
11
|
Fu Q, Polanco A, Lee YS, Yoon S. Critical challenges and advances in recombinant adeno-associated virus (rAAV) biomanufacturing. Biotechnol Bioeng 2023; 120:2601-2621. [PMID: 37126355 DOI: 10.1002/bit.28412] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Gene therapy is a promising therapeutic approach for genetic and acquired diseases nowadays. Among DNA delivery vectors, recombinant adeno-associated virus (rAAV) is one of the most effective and safest vectors used in commercial drugs and clinical trials. However, the current yield of rAAV biomanufacturing lags behind the necessary dosages for clinical and commercial use, which embodies a concentrated reflection of low productivity of rAAV from host cells, difficult scalability of the rAAV-producing bioprocess, and high levels of impurities materialized during production. Those issues directly impact the price of gene therapy medicine in the market, limiting most patients' access to gene therapy. In this context, the current practices and several critical challenges associated with rAAV gene therapy bioprocesses are reviewed, followed by a discussion of recent advances in rAAV-mediated gene therapy and other therapeutic biological fields that could improve biomanufacturing if these advances are integrated effectively into the current systems. This review aims to provide the current state-of-the-art technology and perspectives to enhance the productivity of rAAV while reducing impurities during production of rAAV.
Collapse
Affiliation(s)
- Qiang Fu
- Department of Biomedical Engineering and Biotechnology, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Ashli Polanco
- Department of Chemical Engineering, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Yong Suk Lee
- Department of Pharmaceutical Sciences, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| |
Collapse
|
12
|
Shama, Mahmood A, Mehmood S, Zhang W. Pathological Effects of SARS-CoV-2 Associated with Hematological Abnormalities. Curr Issues Mol Biol 2023; 45:7161-7182. [PMID: 37754237 PMCID: PMC10528388 DOI: 10.3390/cimb45090453] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/09/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
The SARS coronavirus 2 (SARS-CoV-2) is the causative agent of the 2019 coronavirus disease (COVID-19) pandemic that has claimed the lives of 6.9 million people and infected over 765 million. It has become a major worldwide health problem and is also known to cause abnormalities in various systems, including the hematologic system. COVID-19 infection primarily affects the lower respiratory tract and can lead to a cascade of events, including a cytokine storm, intravascular thrombosis, and subsequent complications such as arterial and venous thromboses. COVID-19 can cause thrombocytopenia, lymphopenia, and neutrophilia, which are associated with worse outcomes. Prophylactic anticoagulation is essential to prevent complications and death rates associated with the virus's effect on the coagulation system. It is crucial to recognize these complications early and promptly start therapeutic anticoagulation to improve patient outcomes. While rare, COVID-19-induced disseminated intravascular coagulation (DIC) exhibits some similarities to DIC induced by sepsis. Lactate dehydrogenase (LDH), D-dimer, ferritin, and C-reactive protein (CRP) biomarkers often increase in serious COVID-19 cases and poor prognosis. Understanding the pathophysiology of the disease and identifying risk factors for adverse outcomes is critical for effective management of COVID-19.
Collapse
Affiliation(s)
- Shama
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang 212013, China (A.M.)
| | - Asif Mahmood
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang 212013, China (A.M.)
- School of Material Science and Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Shahid Mehmood
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China;
| | - Wen Zhang
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang 212013, China (A.M.)
| |
Collapse
|
13
|
SARS-CoV-2 Vaccine-Induced T-Cell Response after Three Doses in People Living with HIV on Antiretroviral Therapy Compared to Seronegative Controls (CTN 328 COVAXHIV Study). Viruses 2023; 15:v15020575. [PMID: 36851789 PMCID: PMC9959053 DOI: 10.3390/v15020575] [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: 01/10/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
People living with HIV (PLWH) may be at risk for poor immunogenicity to certain vaccines, including the ability to develop immunological memory. Here, we assessed T-cell immunogenicity following three SARS-CoV-2 vaccine doses in PLWH versus uninfected controls. Blood was collected from 38 PLWH on antiretroviral therapy and 24 age-matched HIV-negative controls, pre-vaccination and after 1st/2nd/3rd dose of SARS-CoV-2 vaccines, without prior SARS-CoV-2 infection. Flow cytometry was used to assess ex vivo T-cell immunophenotypes and intracellular Tumor necrosis factor (TNF)-α/interferon(IFN)-γ/interleukin(IL)-2 following SARS-CoV-2-Spike-peptide stimulation. Comparisons were made using Wilcoxon signed-rank test for paired variables and Mann-Whitney for unpaired. In PLWH, Spike-specific CD4 T-cell frequencies plateaued post-2nd dose, with no significant differences in polyfunctional SARS-CoV-2-specific T-cell proportions between PLWH and uninfected controls post-3rd dose. PLWH had higher frequencies of TNFα+CD4 T-cells and lower frequencies of IFNγ+CD8 T-cells than seronegative participants post-3rd dose. Regardless of HIV status, an increase in naive, regulatory, and PD1+ T-cell frequencies was observed post-3rd dose. In summary, two doses of SARS-CoV-2 vaccine induced a robust T-cell immune response in PLWH, which was maintained after the 3rd dose, with no significant differences in polyfunctional SARS-CoV-2-specific T-cell proportions between PLWH and uninfected controls post-3rd dose.
Collapse
|
14
|
Jergović M, Watanabe M, Bhat R, Coplen CP, Sonar SA, Wong R, Castaneda Y, Davidson L, Kala M, Wilson RC, Twigg HL, Knox K, Erickson HE, Weinkauf CC, Bime C, Bixby BA, Parthasarathy S, Mosier JM, LaFleur BJ, Bhattacharya D, Nikolich JZ. T-cell cellular stress and reticulocyte signatures, but not loss of naïve T lymphocytes, characterize severe COVID-19 in older adults. GeroScience 2023:10.1007/s11357-022-00724-y. [PMID: 36633825 PMCID: PMC9838276 DOI: 10.1007/s11357-022-00724-y] [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: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
In children and younger adults up to 39 years of age, SARS-CoV-2 usually elicits mild symptoms that resemble the common cold. Disease severity increases with age starting at 30 and reaches astounding mortality rates that are ~330 fold higher in persons above 85 years of age compared to those 18-39 years old. To understand age-specific immune pathobiology of COVID-19, we have analyzed soluble mediators, cellular phenotypes, and transcriptome from over 80 COVID-19 patients of varying ages and disease severity, carefully controlling for age as a variable. We found that reticulocyte numbers and peripheral blood transcriptional signatures robustly correlated with disease severity. By contrast, decreased numbers and proportion of naïve T-cells, reported previously as a COVID-19 severity risk factor, were found to be general features of aging and not of COVID-19 severity, as they readily occurred in older participants experiencing only mild or no disease at all. Single-cell transcriptional signatures across age and severity groups showed that severe but not moderate/mild COVID-19 causes cell stress response in different T-cell populations, and some of that stress was unique to old severe participants, suggesting that in severe disease of older adults, these defenders of the organism may be disabled from performing immune protection. These findings shed new light on interactions between age and disease severity in COVID-19.
Collapse
Affiliation(s)
- Mladen Jergović
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Ruchika Bhat
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Christopher P Coplen
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Sandip A Sonar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Rachel Wong
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Vir, Inc., CA, San Francisco, USA
| | - Yvonne Castaneda
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Lisa Davidson
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Mrinalini Kala
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Rachel C Wilson
- Division of Pulmonary Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Homer L Twigg
- Division of Pulmonary Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Kenneth Knox
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Heidi E Erickson
- Department of Medicine, Arizona Respiratory Center, Tucson, AZ, USA
| | - Craig C Weinkauf
- The Division of Vascular Surgery, University of Arizona, Tucson, AZ, USA
| | - Christian Bime
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Billie A Bixby
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Sairam Parthasarathy
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Jarrod M Mosier
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
- Department of Emergency Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Bonnie J LaFleur
- BIO5 Institute, University of Arizona, Tucson, USA
- R. Ken Coit College of Pharmacy, Tucson, AZ, USA
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA
- R. Ken Coit College of Pharmacy, Tucson, AZ, USA
| | - Janko Z Nikolich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, P.O. Box 245221, 1501 N. Campbell Ave, Tucson, AZ, USA.
- Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- R. Ken Coit College of Pharmacy, Tucson, AZ, USA.
| |
Collapse
|
15
|
Aktar S, Amin S. SARS-CoV-2 mediated dysregulation in cell signaling events drives the severity of COVID-19. Virus Res 2023; 323:198962. [PMID: 36209917 PMCID: PMC9536871 DOI: 10.1016/j.virusres.2022.198962] [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] [Received: 03/06/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 01/25/2023]
Abstract
A balance in immune response against an unfamiliar pathogen is crucial to eliminate the infection. A cascade of cell signaling events is immediately activated upon sensing the presence of SARS-CoV-2 by cellular toll like receptors in a natural host response manner against the invading virus. The ultimate aim of such innate immune signaling pathways is to provide a required level of protection to our bodies by interfering with the invader. However, if there is any loss in such balance, an impairment in immune system emerge that fails to control the regulated transcription and translation of signaling components. Consequently, excessive level of proinflammatory mediators release into the circulatory systems that ultimately cause "cytokine storm" and COVID-19 pathological syndromes. The limited production of interferons (IFNs), while excessive yield of pro-inflammatory cytokines followed by SARS-CoV-2 infection suggests an abnormal cell signaling event and explains the reasons of increased immunopathology and severity in COVID-19.
Collapse
Affiliation(s)
- Salma Aktar
- Department of Microbiology, Noakhali Science and Technology University, Noakhali 3814, Bangladesh.
| | - Saiful Amin
- Chittagong Medical University, Chattogram, Bangladesh
| |
Collapse
|
16
|
Porter A, Brown CC, Rodriguez A, Zohoori N, Wells S, Crump A, Romero J, Tilford JM. Variation in Time Between Testing Positive for COVID-19 and Hospital Admission by Race/Ethnicity and Insurance Status. J Health Care Poor Underserved 2023; 34:1290-1304. [PMID: 38661756 PMCID: PMC11101062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Understanding the extent to which demographic and socioeconomic factors play a role in the disparities associated with duration between testing positive for COVID-19 and hospital admission will help in achieving equitable health outcomes. This project linked the statewide COVID-19 registry to administrative datasets to examine the variation in times between testing positive for COVID-19 and hospital admission by race/ethnicity and insurance. In 2020, there were 11,314 patients admitted for COVID-19 in Arkansas. Approximately 42.2% tested positive for COVID-19 on the same day as hospital admission. Black patients had 38% higher odds of hospitalization on the day of testing compared with White patients (p<.001). Medicaid and uninsured patients had 51% and 50% higher odds of admission on the day of testing compared with privately insured patients (both p<.001), respectively. This study highlights the implications of reduced access to testing with respect to equitable health outcomes.
Collapse
Affiliation(s)
- Austin Porter
- Fay W. Boozman College of Public Health, Department of Health Policy and Management, University of Arkansas for Medical Sciences, Little Rock, AR 72205
- Arkansas Department of Health, Little Rock, AR 72205
| | - Clare C. Brown
- Fay W. Boozman College of Public Health, Department of Health Policy and Management, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Namvar Zohoori
- Arkansas Department of Health, Little Rock, AR 72205
- Fay W. Boozman College of Public Health, Department of Epidemiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Samantha Wells
- Fay W. Boozman College of Public Health, Department of Health Policy and Management, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Alisha Crump
- Fay W. Boozman College of Public Health, Department of Epidemiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - José Romero
- Arkansas Department of Health, Little Rock, AR 72205
| | - J. Mick Tilford
- Fay W. Boozman College of Public Health, Department of Health Policy and Management, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| |
Collapse
|
17
|
Abplanalp WT, Merten M, Dimmeler S. Straight to the Heart: T Cells That Specifically Target Cardiac Tissue. Circulation 2022; 146:1946-1949. [PMID: 36534731 DOI: 10.1161/circulationaha.122.061324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Wesley T Abplanalp
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine (W.T.A., M.M., S.D.).,Cardiopulmonary Institute (W.T.A., M.M., S.D.).,Goethe University Frankfurt, Germany (W.T.A., M.M., S.D.).,German Center for Cardiovascular Research DZHK, Partner site Frankfurt Rhine-Main, Berlin, Germany (W.T.A., M.M., S.D.)
| | - Maximilian Merten
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine (W.T.A., M.M., S.D.).,Cardiopulmonary Institute (W.T.A., M.M., S.D.).,Goethe University Frankfurt, Germany (W.T.A., M.M., S.D.).,German Center for Cardiovascular Research DZHK, Partner site Frankfurt Rhine-Main, Berlin, Germany (W.T.A., M.M., S.D.)
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine (W.T.A., M.M., S.D.).,Cardiopulmonary Institute (W.T.A., M.M., S.D.).,Goethe University Frankfurt, Germany (W.T.A., M.M., S.D.).,German Center for Cardiovascular Research DZHK, Partner site Frankfurt Rhine-Main, Berlin, Germany (W.T.A., M.M., S.D.)
| |
Collapse
|
18
|
Abstract
Coronavirus disease 2019 (COVID-19)-associated invasive fungal infections are an important complication in a substantial number of critically ill, hospitalized patients with COVID-19. Three groups of fungal pathogens cause co-infections in COVID-19: Aspergillus, Mucorales and Candida species, including Candida auris. Here we review the incidence of COVID-19-associated invasive fungal infections caused by these fungi in low-, middle- and high-income countries. By evaluating the epidemiology, clinical risk factors, predisposing features of the host environment and immunological mechanisms that underlie the pathogenesis of these co-infections, we set the scene for future research and development of clinical guidance. Hoenigl and colleagues review the epidemiology, immunology and clinical risk factors contributing to COVID-19-associated fungal infections.
Collapse
|
19
|
Attia MH. A cautionary note on altered pace of aging in the COVID-19 era. Forensic Sci Int Genet 2022; 59:102724. [PMID: 35598567 PMCID: PMC9112667 DOI: 10.1016/j.fsigen.2022.102724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/18/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is highly age-dependent due to hi-jacking the molecular control of the immune cells by the severe acute respiratory syndrome-corona virus 2 (SARS-CoV-2) leading to aberrant DNA methylation (DNAm) pattern of blood in comparison to normal individuals. These epigenetic modifications have been linked to perturbations to the epigenetic clock, development of long COVID-19 syndrome, and all-cause mortality risk. I reviewed the effects of COVID-19 on different molecular age markers such as the DNAm, telomere length (TL), and signal joint T-cell receptor excision circle (sjTREC). Integrating the accumulated clinical research data, COVID-19 and novel medical management may alter the pace of aging in adult individuals (<60 years). As such, COVID-19 might be a confounder in epigenetic age estimation similar to life style diversities, pathogens and pathologies which may influence the interpretation of DNAm data. Similarly, the SARS-CoV-2 affects T-lymphocyte function with possible influence on sjTREC levels. In contrast, TL measurements performed years before the SARS-CoV-2 pandemic proved that short TL predisposes to severe COVID- 19 independently from chronological age. However, the persistence of COVID-19 epigenetic scars and the durability of the immune response after vaccination and their effect on the ongoing pace of aging are still unknown. In the light of these data, the heterogeneous nature of the samples in these studies mandates a systematic evaluation of the currrent methods. SARS-CoV-2 may modify the reliability of the age estimation models in real casework because blood is the most common biological sample encountered in forensic contexts.
Collapse
|
20
|
Hoenigl M, Seidel D, Carvalho A, Rudramurthy SM, Arastehfar A, Gangneux JP, Nasir N, Bonifaz A, Araiza J, Klimko N, Serris A, Lagrou K, Meis JF, Cornely OA, Perfect JR, White PL, Chakrabarti A. The emergence of COVID-19 associated mucormycosis: a review of cases from 18 countries. THE LANCET. MICROBE 2022; 3:e543-e552. [PMID: 35098179 PMCID: PMC8789240 DOI: 10.1016/s2666-5247(21)00237-8] [Citation(s) in RCA: 259] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Reports of COVID-19-associated mucormycosis have been increasing in frequency since early 2021, particularly among patients with uncontrolled diabetes. Patients with diabetes and hyperglycaemia often have an inflammatory state that could be potentiated by the activation of antiviral immunity to SARS-CoV2, which might favour secondary infections. In this Review, we analysed 80 published and unpublished cases of COVID-19-associated mucormycosis. Uncontrolled diabetes, as well as systemic corticosteroid treatment, were present in most patients with COVID-19-associated mucormycosis, and rhino-orbital cerebral mucormycosis was the most frequent disease. Mortality was high at 49%, which was particularly due to patients with pulmonary or disseminated mucormycosis or cerebral involvement. Furthermore, a substantial proportion of patients who survived had life-changing morbidities (eg, loss of vision in 46% of survivors). Our Review indicates that COVID-19-associated mucormycosis is associated with high morbidity and mortality. Diagnosis of pulmonary mucormycosis is particularly challenging, and might be frequently missed in India.
Collapse
Affiliation(s)
- Martin Hoenigl
- Division of Infectious Diseases, ECMM Center of Excellence for Medical Mycology, Medical University of Graz, Graz, Austria
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Clinical and Translational Fungal Working Group, University of California San Diego, La Jolla, CA, USA
| | - Danila Seidel
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Department of Internal Medicine, ECMM Center of Excellence for Medical Mycology, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- PT Government Associate Laboratory, Guimarães, Portugal
| | - Shivaprakash M Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amir Arastehfar
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Jean-Pierre Gangneux
- Environnement et Travail, Univ Rennes, CHU Rennes, Inserm, Institut de Recherche en Santé, Rennes, France
| | - Nosheen Nasir
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University Karachi, Karachi, Pakistan
| | - Alexandro Bonifaz
- Dermatology Service, Hospital General De México Dr Eduardo Liceaga, Mexico City, Mexico
| | - Javier Araiza
- Dermatology Service, Hospital General De México Dr Eduardo Liceaga, Mexico City, Mexico
| | - Nikolai Klimko
- Department of Clinical Mycology, Allergy and Immunology, North Western State Medical University named after II Mechnikov, St Petersburg, Russia
| | - Alexandra Serris
- Department of Infectious Diseases, Necker-Enfants Malades University Hospital, Paris, France
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine and National Reference Centre for Mycosis, ECMM Center of Excellence for Medical Mycology, University Hospitals Leuven, Leuven, Belgium
| | - Jacques F Meis
- Department of Medical Microbiology and Infectious Diseases, ECMM Center of Excellence for Medical Mycology, Radboud University Medical Center, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
- Center of Expertise in Mycology, Radboud University Medical Center, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
- Bioprocess Engineering and Biotechnology Graduate Program, Federal University of Paraná, Curitiba, Brazil
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Department of Internal Medicine, ECMM Center of Excellence for Medical Mycology, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Clinical Trials Centre Cologne, ZKS Köln, University of Cologne, Cologne, Germany
| | - John R Perfect
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - P Lewis White
- Public Health Wales Mycology Reference Laboratory, UHW, Cardiff, UK
| | - Arunaloke Chakrabarti
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
21
|
Savchenko AA, Tikhonova E, Kudryavtsev I, Kudlay D, Korsunsky I, Beleniuk V, Borisov A. TREC/KREC Levels and T and B Lymphocyte Subpopulations in COVID-19 Patients at Different Stages of the Disease. Viruses 2022; 14:646. [PMID: 35337053 PMCID: PMC8954181 DOI: 10.3390/v14030646] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND T and B cell-mediated immunity can be assessed using T cell receptor excision circle (TREC) and Kappa-deleting recombination excision circle (KREC) analysis, respectively, and successful implementation of this method requires evaluation of the correlation between the TREC frequencies and T cell subsets as well as KREC levels and B lymphocyte subsets. The aim of the present study was to evaluate the correlation between the TREC/KREC concentrations and T/B lymphocyte subsets at different stages of COVID-19. METHODS We examined 33 patients in the acute stage of COVID-19 (including 8 patients with poor outcomes) and 33 COVID-19 survivors. TREC/KREC concentrations were measured using quantitative real-time PCR. T/B lymphocyte subsets were determined using flow cytometry. RESULTS Blood TREC and KREC levels were found to be significantly lower in the acute stage of COVID-19 compared to control values. Moreover, a zero blood TREC level was a predictor of a poor disease outcome. Reductions in CD3+CD4+CD45RO-CD62L- and CD3+CD8+CD45RO-CD62L- T cell counts (as well as in the main fractions of B1 and B2 B cells) indicated a favorable outcome in COVID-19 patients in the acute stage of the disease. Decreased CD3+CD4+CD45RO-CD62L+ and CD3+CD8+CD45RO-CD62L+ T cell frequencies and increased CD3+CD8+CD45RO-CD62L- cell counts were found to indicate a poor outcome in patients with acute COVID-19. These patients were also found to have increased B1 cell counts while demonstrating no changes in B2 cell counts. The levels of effector T cell subsets an naïve B cells were normal in COVID-19 survivors. The most pronounced correlations between TREC/KREC levels and T/B cell subsets counts were observed in COVID-19 survivors: there were positive correlations with naïve T and B lymphocytes and negative correlations with central and effector memory T cell subsets. CONCLUSIONS The assessment of correlations between TREC and T cell subsets as well as KREC levels and B cell subset counts in patients with acute COVID-19 and COVID-19 survivors has shown that blood concentrations of TREC and KREC are sensitive indicators of the stage of antigen-independent differentiation of adaptive immunity cells. The results of the TREC and KREC analysis correlated with the stages of COVID-19 and differed depending on the outcome of COVID-19.
Collapse
Affiliation(s)
- Andrei A. Savchenko
- Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.A.S.); (V.B.); (A.B.)
| | - Elena Tikhonova
- Ministry of Health of the Russian Federation, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia;
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Dmitry Kudlay
- National Research Center—Institute of Immunology, Federal Medical-Biological Agency, 115522 Moscow, Russia;
- Ministry of Health of the Russian Federation, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Ilya Korsunsky
- Moscow City Center for Pediatric Immunology and Allergy, G. Speransky Children’s Hospital No 9, 129329 Moscow, Russia;
| | - Vasily Beleniuk
- Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.A.S.); (V.B.); (A.B.)
| | - Alexandr Borisov
- Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia; (A.A.S.); (V.B.); (A.B.)
| |
Collapse
|
22
|
Chakrabarti SS, Kaur U, Aggarwal SK, Kanakan A, Saini A, Agrawal BK, Jin K, Chakrabarti S. The Pathogenetic Dilemma of Post-COVID-19 Mucormycosis in India. Aging Dis 2022; 13:24-28. [PMID: 35111359 PMCID: PMC8782544 DOI: 10.14336/ad.2021.0811] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022] Open
Abstract
There has been a surge of mucormycosis cases in India in the wake of the second wave of COVID-19 with more than 40000 cases reported. Mucormycosis in patients of COVID-19 in India is at variance to other countries where Aspergillus, Pneumocystis, and Candida have been reported to be the major secondary fungal pathogens. We discuss the probable causes of the mucormycosis epidemic in India. Whereas dysglycaemia and inappropriate steroid use have been widely suggested as tentative reasons, we explore other biological, iatrogenic, and environmental factors. The likelihood of a two-hit pathogenesis remains strong. We propose that COVID-19 itself provides the predisposition to invasive mucormycosis (first hit), through upregulation of GRP78 and downregulation of spleen tyrosine kinase involved in anti-fungal defense, as also through inhibition of CD8+ T-cell mediated immunity. The other iatrogenic and environmental factors may provide the second hit which may have resulted in the surge.
Collapse
Affiliation(s)
- Sankha Shubhra Chakrabarti
- 1Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India
| | - Upinder Kaur
- 2Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India
| | - Sushil Kumar Aggarwal
- 3Department of Otorhinolaryngology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India
| | - Ahalya Kanakan
- 4Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India
| | - Adesh Saini
- 5Department of Biotechnology, Maharishi Markandeshwar (deemed to be) University, Mullana, Haryana, India
| | - Bimal Kumar Agrawal
- 6Department of Medicine, Maharishi Markandeshwar (deemed to be) University, Mullana, Haryana, India
| | - Kunlin Jin
- 7Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Sasanka Chakrabarti
- 8Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar (deemed to be) University, Mullana, Haryana, India
| |
Collapse
|
23
|
Wang S, Hao M, Pan Z, Lei J, Zou X. Data-driven multi-scale mathematical modeling of SARS-CoV-2 infection reveals heterogeneity among COVID-19 patients. PLoS Comput Biol 2021; 17:e1009587. [PMID: 34818337 PMCID: PMC8654229 DOI: 10.1371/journal.pcbi.1009587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 12/08/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) often exhibit diverse disease progressions associated with various infectious ability, symptoms, and clinical treatments. To systematically and thoroughly understand the heterogeneous progression of COVID-19, we developed a multi-scale computational model to quantitatively understand the heterogeneous progression of COVID-19 patients infected with severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2). The model consists of intracellular viral dynamics, multicellular infection process, and immune responses, and was formulated using a combination of differential equations and stochastic modeling. By integrating multi-source clinical data with model analysis, we quantified individual heterogeneity using two indexes, i.e., the ratio of infected cells and incubation period. Specifically, our simulations revealed that increasing the host antiviral state or virus induced type I interferon (IFN) production rate can prolong the incubation period and postpone the transition from asymptomatic to symptomatic outcomes. We further identified the threshold dynamics of T cell exhaustion in the transition between mild-moderate and severe symptoms, and that patients with severe symptoms exhibited a lack of naïve T cells at a late stage. In addition, we quantified the efficacy of treating COVID-19 patients and investigated the effects of various therapeutic strategies. Simulations results suggested that single antiviral therapy is sufficient for moderate patients, while combination therapies and prevention of T cell exhaustion are needed for severe patients. These results highlight the critical roles of IFN and T cell responses in regulating the stage transition during COVID-19 progression. Our study reveals a quantitative relationship underpinning the heterogeneity of transition stage during COVID-19 progression and can provide a potential guidance for personalized therapy in COVID-19 patients. Coronavirus disease 2019 (COVID-19) is currently destroying both lives and economies. However, patients infected with severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) usually present heterogeneous and complicated progressions, such as different incubation periods (short and long), symptoms (asymptomatic and symptomatic) and severity (mild-moderate and severe). Currently, various clinical data and experimental data are available from different countries, which has great significance for integrating different types of data to comprehensively understand the diverse disease progression in COVID-19 patients and guide individual treatment strategies. Here, we developed a multi-scale computational model to describe the dynamical process of patients infected with SARS-CoV-2, including intracellular viral dynamics, multicellular infection process, and immune responses. By combining data integration, stochastic simulation and quantitative analysis based on the multi-scale mathematical model, we addressed an important question regarding how IFN response and T cell exhaustion quantitatively affect heterogeneous progression in patients with respect to incubation periods, symptoms and severity. Furthermore, the efficacy of various therapeutic strategies for treating COVID-19 patients with different severity degrees was evaluated and validated. The computational framework in this study can also be extended to explore the dynamical process of other coronavirus infections.
Collapse
Affiliation(s)
- Shun Wang
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
| | - Mengqian Hao
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
| | - Zishu Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jinzhi Lei
- School of Mathematical Sciences, Center for Applied Mathematics, Tiangong University, Tianjin, China
- * E-mail: (JL); (XZ)
| | - Xiufen Zou
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
- * E-mail: (JL); (XZ)
| |
Collapse
|
24
|
Karim MM, Sultana S, Sultana R, Rahman MT. Possible Benefits of Zinc supplement in CVD and COVID-19 Comorbidity. J Infect Public Health 2021; 14:1686-1692. [PMID: 34649043 PMCID: PMC8489295 DOI: 10.1016/j.jiph.2021.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 09/09/2021] [Accepted: 09/29/2021] [Indexed: 12/31/2022] Open
Abstract
As far as comorbidity is concerned, cardiovascular diseases (CVD) appear to be accounted for the highest prevalence, severity, and fatality among COVID 19 patients. A wide array of causal links connecting CVD and COVID-19 baffle the overall prognosis as well as the efficacy of the given therapeutic interventions. At the centre of this puzzle lies ACE2 that works as a receptor for the SARS-CoV-2, and functional expression of which is also needed to minimize vasoconstriction otherwise would lead to high blood pressure. Furthermore, SARS-CoV-2 infection seems to reduce the functional expression of ACE2. Given these circumstances, it might be advisable to consider a treatment plan for COVID-19 patients with CVD in an approach that would neither aggravate the vasodeleterious arm of the renin-angiotensinogen-aldosterone system (RAAS) nor compromise the vasoprotective arm of RAAS but is effective to minimize or if possible, inhibit the viral replication. Given the immune modulatory role of Zn in both CVD and COVID-19 pathogenesis, zinc supplement to the selective treatment plan for CVD and COVID-19 comorbid conditions, to be decided by the clinicians depending on the cardiovascular conditions of the patients, might greatly improve the therapeutic outcome. Notably, ACE2 is a zinc metalloenzyme and zinc is also known to inhibit viral replication.
Collapse
Affiliation(s)
| | - Shahnaz Sultana
- Institute of National Analytical Research and Service (INARS), Bangladesh Council of Scientific and Industrial Research (BCSIR), New Elephant Road, Dhaka 1205, Bangladesh
| | - Rokaia Sultana
- Institute of National Analytical Research and Service (INARS), Bangladesh Council of Scientific and Industrial Research (BCSIR), New Elephant Road, Dhaka 1205, Bangladesh
| | - Mohammad Tariqur Rahman
- Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia,Corresponding author
| |
Collapse
|
25
|
Yan T, Xiao R, Wang N, Shang R, Lin G. Obesity and severe coronavirus disease 2019: molecular mechanisms, paths forward, and therapeutic opportunities. Theranostics 2021; 11:8234-8253. [PMID: 34373739 PMCID: PMC8343994 DOI: 10.7150/thno.59293] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/20/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) appears to have higher pathogenicity among patients with obesity. Obesity, termed as body mass index greater than 30 kg/m2, has now been demonstrated to be important comorbidity for disease severity during coronavirus disease 2019 (COVID-19) pandemic and associated with adverse events. Unraveling mechanisms behind this phenomenon can assist scientists, clinicians, and policymakers in responding appropriately to the COVID-19 pandemic. In this review, we systemically delineated the potential mechanistic links between obesity and worsening COVID-19 from altered physiology, underlying diseases, metabolism, immunity, cytokine storm, and thrombosis. Problematic ventilation caused by obesity and preexisting medical disorders exacerbate organ dysfunction for patients with obesity. Chronic metabolic disorders, including dyslipidemia, hyperglycemia, vitamin D deficiency, and polymorphisms of metabolism-related genes in obesity, probably aid SARS-CoV-2 intrusion and impair antiviral responses. Obesity-induced inadequate antiviral immunity (interferon, natural killer cells, invariant natural killer T cell, dendritic cell, T cells, B cell) at the early stage of SARS-CoV-2 infection leads to delayed viral elimination, increased viral load, and expedited viral mutation. Cytokine storm, with the defective antiviral immunity, probably contributes to tissue damage and pathological progression, resulting in severe symptoms and poor prognosis. The prothrombotic state, driven in large part by endothelial dysfunction, platelet hyperactivation, hypercoagulability, and impaired fibrinolysis in obesity, also increases the risk of severe COVID-19. These mechanisms in the susceptibility to severe condition also open the possibility for host-directed therapies in population with obesity. By bridging work done in these fields, researchers can gain a holistic view of the paths forward and therapeutic opportunities to break the vicious cycle of obesity and its devastating complications in the next emerging pandemic.
Collapse
Affiliation(s)
- Tiantian Yan
- Military Burn Center, the 990th Hospital of People's Liberation Army Joint Logistics Support Force, Zhumadian, Henan, China
| | - Rong Xiao
- Military Burn Center, the 990th Hospital of People's Liberation Army Joint Logistics Support Force, Zhumadian, Henan, China
| | - Nannan Wang
- Military Burn Center, the 990th Hospital of People's Liberation Army Joint Logistics Support Force, Zhumadian, Henan, China
| | - Ruoyu Shang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Guoan Lin
- Military Burn Center, the 990th Hospital of People's Liberation Army Joint Logistics Support Force, Zhumadian, Henan, China
| |
Collapse
|
26
|
Xie C, Li Q, Li L, Peng X, Ling Z, Xiao B, Feng J, Chen Z, Chang D, Xie L, Dela Cruz CS, Sharma L. Association of Early Inflammation with Age and Asymptomatic Disease in COVID-19. J Inflamm Res 2021; 14:1207-1216. [PMID: 33833543 PMCID: PMC8020811 DOI: 10.2147/jir.s304190] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Disease severity in COVID-19 ranges from asymptomatic infection to severe disease and death, especially in older subjects. The risk for severe infection and death has been reported to be 2X in those between 30 and 40 years, 3X in those between 40 and 50 years, and 4X in those between 50 and 65 years, compared to the reference group of 18-29 years. OBJECTIVE To investigate the early changes in host immune responses that are altered with age and the difference in the early host inflammatory response that dictates a symptomatic versus asymptomatic course of COVID-19. PATIENTS AND METHODS COVID-19 subjects were identified by screening at the airport upon arrival from a foreign destination to China. Patients were either asymptomatic or had a mild disease when the first oro-pharyngeal (OP) swab samples were collected. Patients were quarantined and blood and throat swabs were collected during the course of the disease, allowing identification of the earliest host response to COVID-19. These patients were followed until their OP sample turned COVID-19 negative. RESULTS Data were obtained from 126 PCR-confirmed COVID-19 patients. The blood samples were obtained within 48 days of qPCR confirmation of viral infection. Older subjects (>30 years) had significantly elevated levels of anti-inflammatory cytokine IL-10, a significant decrease in the percentage of CD8+ T cells, and expansion in NKT cell fraction. This was associated with significantly elevated viral load and a delayed humoral response in older subjects. Compared to symptomatic subjects, asymptomatic patients had an early increase in pro-inflammatory cytokine IL-2, while a decrease in both T regulatory cells and anti-inflammatory cytokine IL-10. Further, asymptomatic disease was associated with early humoral response and faster viral clearance. CONCLUSION Early inflammatory response potentially plays a critical role for host-defense in COVID-19. The impaired early inflammatory response was associated with older age while a robust early inflammation was associated with asymptomatic disease.
Collapse
Affiliation(s)
- Chunmei Xie
- Department of Blood Transfusion, Guangzhou 8th People’s Hospital, Guangzhou Medical University, Guangzhou, 510440, People’s Republic of China
| | - Qing Li
- Department of Blood Transfusion, Guangzhou 8th People’s Hospital, Guangzhou Medical University, Guangzhou, 510440, People’s Republic of China
| | - Linhai Li
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100083, People’s Republic of China
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, People’s Republic of China
| | - Xiaohua Peng
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhijian Ling
- Department of Blood Transfusion, Guangzhou 8th People’s Hospital, Guangzhou Medical University, Guangzhou, 510440, People’s Republic of China
| | - Bin Xiao
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, 510010, People’s Republic of China
| | - Jingjing Feng
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, People’s Republic of China
| | - Zhenhong Chen
- Department of Pulmonary and Critical Care Medicine, Secondary Medical Center, Chinese PLA General Hospital, Beijing, 100083, People’s Republic of China
| | - De Chang
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100083, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Third Medical Center, Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100083, People’s Republic of China
| | - Charles S Dela Cruz
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Lokesh Sharma
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
27
|
Engin AB, Engin ED, Engin A. The effect of environmental pollution on immune evasion checkpoints of SARS-CoV-2. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 81:103520. [PMID: 33132153 PMCID: PMC7580701 DOI: 10.1016/j.etap.2020.103520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 05/04/2023]
Abstract
Many diverse strategies allow and facilitate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to evade antiviral innate immune mechanisms. Although the type I interferon (IFN) system has a critical role in restricting the dissemination of viral infection, suppression of IFN receptor signals by SARS-CoV-2 constitutes a checkpoint that plays an important role in the immune escape of the virus. Environmental pollution not only facilitates SARS-CoV-2 infection but also increases infection-associated fatality risk, which arises due to Systemic Aryl hydrocarbon Receptor (AhR) Activation Syndrome. The intracellular accumulation of endogenous kynurenic acid due to overexpression of the indoleamine 2,3-dioxygenase (IDO) by AhR activation induces AhR-interleukin-6 (IL-6)-signal transducers and activators of the transcription 3 (STAT3) signaling pathway. The AhR-IDO1-Kynurenine pathway is an important checkpoint, which leads to fatal consequences in SARS-CoV-2 infection and immune evasion in the context of Treg/Th17 imbalance and cytokine storm.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey.
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Gumusdere Campus, Kecioren, Ankara, Turkey
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| |
Collapse
|
28
|
Zöphel D, Hof C, Lis A. Altered Ca 2+ Homeostasis in Immune Cells during Aging: Role of Ion Channels. Int J Mol Sci 2020; 22:ijms22010110. [PMID: 33374304 PMCID: PMC7794837 DOI: 10.3390/ijms22010110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Aging is an unstoppable process and begins shortly after birth. Each cell of the organism is affected by the irreversible process, not only with equal density but also at varying ages and with different speed. Therefore, aging can also be understood as an adaptation to a continually changing cellular environment. One of these very prominent changes in age affects Ca2+ signaling. Especially immune cells highly rely on Ca2+-dependent processes and a strictly regulated Ca2+ homeostasis. The intricate patterns of impaired immune cell function may represent a deficit or compensatory mechanisms. Besides, altered immune function through Ca2+ signaling can profoundly affect the development of age-related disease. This review attempts to summarize changes in Ca2+ signaling due to channels and receptors in T cells and beyond in the context of aging.
Collapse
Affiliation(s)
| | | | - Annette Lis
- Correspondence: ; Tel.: +49-(0)-06841-1616318; Fax: +49-(0)-6841-1616302
| |
Collapse
|