1
|
Lu Y, Wang Y, Ruan T, Wang Y, Ju L, Zhou M, Liu L, Yao D, Yao M. Immunometabolism of Tregs: mechanisms, adaptability, and therapeutic implications in diseases. Front Immunol 2025; 16:1536020. [PMID: 39917294 PMCID: PMC11798928 DOI: 10.3389/fimmu.2025.1536020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Immunometabolism is an emerging field that explores the intricate interplay between immune cells and metabolism. Regulatory T cells (Tregs), which maintain immune homeostasis in immunometabolism, play crucial regulatory roles. The activation, differentiation, and function of Tregs are influenced by various metabolic pathways, such as the Mammalian targets of rapamycin (mTOR) pathway and glycolysis. Correspondingly, activated Tregs can reciprocally impact these metabolic pathways. Tregs also possess robust adaptive capabilities, thus enabling them to adapt to various microenvironments, including the tumor microenvironment (TME). The complex mechanisms of Tregs in metabolic diseases are intriguing, particularly in conditions like MASLD, where Tregs are significantly upregulated and contribute to fibrosis, while in diabetes, systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA), they show downregulation and reduced anti-inflammatory capacity. These phenomena suggest that the differentiation and function of Tregs are influenced by the metabolic environment, and imbalances in either can lead to the development of metabolic diseases. Thus, moderate differentiation and inhibitory capacity of Tregs are critical for maintaining immune system balance. Given the unique immunoregulatory abilities of Tregs, the development of targeted therapeutic drugs may position them as novel targets in immunotherapy. This could contribute to restoring immune system balance, resolving metabolic dysregulation, and fostering innovation and progress in immunotherapy.
Collapse
|
2
|
Song S, Gan J, Long Q, Gao Z, Zheng Y. Decoding NAD+ Metabolism in COVID-19: Implications for Immune Modulation and Therapy. Vaccines (Basel) 2024; 13:1. [PMID: 39852780 PMCID: PMC11768799 DOI: 10.3390/vaccines13010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025] Open
Abstract
The persistent threat of COVID-19, particularly with the emergence of new variants, underscores the urgency for innovative therapeutic strategies beyond conventional antiviral treatments. Current immunotherapies, including IL-6/IL-6R monoclonal antibodies and JAK inhibitors, exhibit suboptimal efficacy, necessitating alternative approaches. Our review delves into the significance of NAD+ metabolism in COVID-19 pathology, marked by decreased NAD+ levels and upregulated NAD+-consuming enzymes such as CD38 and poly (ADP-ribose) polymerases (PARPs). Recognizing NAD+'s pivotal role in energy metabolism and immune modulation, we propose modulating NAD+ homeostasis could bolster the host's defensive capabilities against the virus. The article reviews the scientific rationale behind targeting NAD+ pathways for therapeutic benefit, utilizing strategies such as NAD+ precursor supplementation and enzyme inhibition to modulate immune function. While preliminary data are encouraging, the challenge lies in optimizing these interventions for clinical use. Future research should aim to unravel the intricate roles of key metabolites and enzymes in NAD+ metabolism and to elucidate their specific mechanisms of action. This will be essential for developing targeted NAD+ therapies, potentially transforming the management of COVID-19 and setting a precedent for addressing other infectious diseases.
Collapse
Affiliation(s)
- Shixu Song
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| | - Jialing Gan
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| | - Qiuyue Long
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| | - Zhancheng Gao
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing 100044, China
| | - Yali Zheng
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Institute of Chest and Lung Diseases, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| |
Collapse
|
3
|
Kocsmár É, Kocsmár I, Elamin F, Pápai L, Jakab Á, Várkonyi T, Glasz T, Rácz G, Pesti A, Danics K, Kiss A, Röst G, Belicza É, Schaff Z, Lotz G. Autopsy findings in cancer patients infected with SARS-CoV-2 show a milder presentation of COVID-19 compared to non-cancer patients. GeroScience 2024; 46:6101-6114. [PMID: 38691298 PMCID: PMC11493920 DOI: 10.1007/s11357-024-01163-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/12/2024] [Indexed: 05/03/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, manifests with differing severity across distinct patient subgroups, with outcomes influenced by underlying comorbidities such as cancer, which may cause functional and compositional alterations of the immune system during tumor progression. We aimed to investigate the association of SARS-CoV-2 infection and its complications with cancer in a large autopsy series and the role of COVID-19 in the fatal sequence leading to death. A total of 2641 adult autopsies were investigated, 539 of these were positive for SARS-CoV-2. Among the total number of patients analyzed, 829 had active cancer. Overall, the cohort included 100 patients who simultaneously had cancer and SARS-CoV-2 infection. The course of COVID-19 was less severe in cancer patients, including a significantly lower incidence of viral and bacterial pneumonia, occurring more frequently as a contributory disease or coexisting morbidity, or as SARS-CoV-2 positivity without viral disease. SARS-CoV-2 positivity was more frequent among non-metastatic than metastatic cancer cases, and in specific tumor types including hematologic malignancies. COVID-19 was more frequently found to be directly involved in the fatal sequence in patients undergoing active anticancer therapy, but less frequently in perioperative status, suggesting that the underlying malignancy and consequent surgery are more important factors leading to death perioperatively than viral disease. The course of COVID-19 in cancer patients was milder and balanced during the pandemic. This may be due to relative immunosuppressed status, and the fact that even early/mild viral infections can easily upset their condition, leading to death from their underlying cancer or its complications.
Collapse
Affiliation(s)
- Éva Kocsmár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| | - Ildikó Kocsmár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Flóra Elamin
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Laura Pápai
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Ákos Jakab
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tibor Várkonyi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tibor Glasz
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Adrián Pesti
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Krisztina Danics
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - András Kiss
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Gergely Röst
- National Laboratory for Health Security, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Éva Belicza
- Health Services Management Training Centre, Faculty of Health and Public Administration, Semmelweis University, Budapest, Hungary
| | - Zsuzsa Schaff
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Gábor Lotz
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
4
|
Shi J, Shen L, Xiao Y, Wan C, Wang B, Zhou P, Zhang J, Han W, Hu R, Yu F, Wang H. Identification and validation of diagnostic biomarkers and immune cell abundance characteristics in Staphylococcus aureus bloodstream infection by integrative bioinformatics analysis. Front Immunol 2024; 15:1450782. [PMID: 39654884 PMCID: PMC11626409 DOI: 10.3389/fimmu.2024.1450782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/01/2024] [Indexed: 12/12/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic pathogen that could cause life-threatening bloodstream infections. The objective of this study was to identify potential diagnostic biomarkers of S. aureus bloodstream infection. Gene expression dataset GSE33341 was optimized as the discovery dataset, which contained samples from human and mice. GSE65088 dataset was utilized as a validation dataset. First, after overlapping the differentially expressed genes (DEGs) in S. aureus infection samples from GSE33341-human and GSE33341-mice samples, we detected 63 overlapping genes. Subsequently, the hub genes including DRAM1, PSTPIP2, and UPP1 were identified via three machine-learning algorithms: random forest, support vector machine-recursive feature elimination, and least absolute shrinkage and selection operator. Additionally, the receiver operating characteristic curve was leveraged to verify the efficacy of the hub genes. DRAM1 (AUC=1), PSTPIP2 (AUC=1), and UPP1 (AUC=1) were investigated and demonstrated significant expression differences (all P < 0.05) and diagnostic efficacy in the training and validation datasets. Furthermore, the relationship between the diagnostic markers and the abundance of immune cells was assessed using cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT). These three diagnostic indicators also correlated with multiple immune cells to varying degrees. The expression of DRAM1 was significantly positively correlated with B cell naive and mast cell activation, and negatively correlated with NK cells and CD4/CD8+ T cells. The expression of PSTPIP2 was significantly positively correlated with macrophage M0, macrophage M1, B cell naive, and dendritic cell activation, while the expression of PSTPIP2 was negatively correlated with NK cells and CD4/CD8+ T cells. Significant negative correlations between UPP1 expression and T cell CD4 memory rest and neutrophils were also observed. Finally, we established a mouse model of S. aureus bloodstream infection and collected the blood samples for RNA-Seq analysis and RT-qPCR experiments. The analysis results in RNA-Seq and RT-qPCR experiments further confirmed the significant expression differences (all P < 0.05) of these three genes. Overall, three candidate hub genes (DRAM1, PSTPIP2, and UPP1) were identified initially for S. aureus bloodstream infection diagnosis. Our study could provide potential diagnostic biomarkers for S. aureus bloodstream infection patients.
Collapse
Affiliation(s)
- Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cailing Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rongrong Hu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongxiu Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Consonni FM, Incerti M, Bertolotti M, Ballerini G, Garlatti V, Sica A. Heme catabolism and heme oxygenase-1-expressing myeloid cells in pathophysiology. Front Immunol 2024; 15:1433113. [PMID: 39611159 PMCID: PMC11604077 DOI: 10.3389/fimmu.2024.1433113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/01/2024] [Indexed: 11/30/2024] Open
Abstract
Although the pathological significance of myeloid cell heterogeneity is still poorly understood, new evidence indicates that distinct macrophage subsets are characterized by specific metabolic programs that influence disease onset and progression. Within this scenario, distinct subsets of macrophages, endowed with high rates of heme catabolism by the stress-responsive enzyme heme oxygenase-1 (HO-1), play critical roles in physiologic and pathological conditions. Of relevance, the substrates of HO-1 activity are the heme groups that derive from cellular catabolism and are converted into carbon monoxide (CO), biliverdin and Fe2+, which together elicit anti-apoptotic, anti-inflammatory activities and control oxidative damage. While high levels of expression of HO-1 enzyme by specialized macrophage populations (erythrophagocytes) guarantee the physiological disposal of senescent red blood cells (i.e. erythrocateresis), the action of HO-1 takes on pathological significance in various diseases, and abnormal CO metabolism has been observed in cancer, hematological diseases, hypertension, heart failure, inflammation, sepsis, neurodegeneration. Modulation of heme catabolism and CO production is therefore a feasible therapeutic opportunity in various diseases. In this review we discuss the role of HO-1 in different pathological contexts (i.e. cancer, infections, cardiovascular, immune-mediated and neurodegenerative diseases) and highlight new therapeutic perspectives on the modulation of the enzymatic activity of HO-1.
Collapse
Affiliation(s)
- Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Martina Incerti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Milena Bertolotti
- Navita S.r.l., University of Eastern Piedmont A. Avogadro, Novara, Italy
| | - Giulia Ballerini
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Valentina Garlatti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
6
|
Jones RP, Ponomarenko A. COVID-19-Related Age Profiles for SARS-CoV-2 Variants in England and Wales and States of the USA (2020 to 2022): Impact on All-Cause Mortality. Infect Dis Rep 2023; 15:600-634. [PMID: 37888139 PMCID: PMC10606787 DOI: 10.3390/idr15050058] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 10/28/2023] Open
Abstract
Since 2020, COVID-19 has caused serious mortality around the world. Given the ambiguity in establishing COVID-19 as the direct cause of death, we first investigate the effects of age and sex on all-cause mortality during 2020 and 2021 in England and Wales. Since infectious agents have their own unique age profile for death, we use a 9-year time series and several different methods to adjust single-year-of-age deaths in England and Wales during 2019 (the pre-COVID-19 base year) to a pathogen-neutral single-year-of-age baseline. This adjusted base year is then used to confirm the widely reported higher deaths in males for most ages above 43 in both 2020 and 2021. During 2020 (+COVID-19 but no vaccination), both male and female population-adjusted deaths significantly increased above age 35. A significant reduction in all-cause mortality among both males and females aged 75+ could be demonstrated in 2021 during the widespread COVID-19 vaccination period; however, deaths below age 75 progressively increased. This finding arises from a mix of vaccination coverage and year-of-age profiles of deaths for the different SARS-CoV-2 variants. In addition, specific effects of age around puberty were demonstrated, where females had higher deaths than males. There is evidence that year-of-birth cohorts may also be involved, indicating that immune priming to specific pathogen outbreaks in the past may have led to lower deaths for some birth cohorts. To specifically identify the age profile for the COVID-19 variants from 2020 to 2023, we employ the proportion of total deaths at each age that are potentially due to or 'with' COVID-19. The original Wuhan strain and the Alpha variant show somewhat limited divergence in the age profile, with the Alpha variant shifting to a moderately higher proportion of deaths below age 84. The Delta variant specifically targeted individuals below age 65. The Omicron variants showed a significantly lower proportion of overall mortality, with a markedly higher relative proportion of deaths above age 65, steeply increasing with age to a maximum around 100 years of age. A similar age profile for the variants can be seen in the age-banded deaths in US states, although they are slightly obscured by using age bands rather than single years of age. However, the US data shows that higher male deaths are greatly dependent on age and the COVID variant. Deaths assessed to be 'due to' COVID-19 (as opposed to 'involving' COVID-19) in England and Wales were especially overestimated in 2021 relative to the change in all-cause mortality. This arose as a by-product of an increase in COVID-19 testing capacity in late 2020. Potential structure-function mechanisms for the age-specificity of SARS-CoV-2 variants are discussed, along with potential roles for small noncoding RNAs (miRNAs). Using data from England, it is possible to show that the unvaccinated do indeed have a unique age profile for death from each variant and that vaccination alters the shape of the age profile in a manner dependent on age, sex, and the variant. The question is posed as to whether vaccines based on different variants carry a specific age profile.
Collapse
Affiliation(s)
| | - Andrey Ponomarenko
- Department of Biophysics, Informatics and Medical Instrumentation, Odessa National Medical University, Valikhovsky Lane 2, 65082 Odessa, Ukraine
| |
Collapse
|
7
|
Zheng A, Huang N, Bean D, Rayapaneni S, Deeney J, Sagar M, Hamilton JA. Resolvin E1 heals injured cardiomyocytes: Therapeutic implications and H-FABP as a readout for cardiovascular disease & systemic inflammation. Prostaglandins Leukot Essent Fatty Acids 2023; 197:102586. [PMID: 37604082 PMCID: PMC11203388 DOI: 10.1016/j.plefa.2023.102586] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023]
Abstract
The purpose of this study is to investigate heart-fatty acid binding protein (H-FABP) leakage from cardiomyocytes as a quantitative measure of cell membrane damage and to test healing by Resolvin E1 (RVE1) as a potential therapeutic for patients with inflammatory diseases (cardiovascular disease and comorbidities) with high morbidity and mortality. Our quantitative ELISA assays demonstrated H-FABP as a sensitive and reliable biomarker for measuring cardiomyocyte damage induced by lipopolysaccharide (LPS) and healing by RvE1, a specialized pro-resolving mediator (SPM) derived from the Omega-3 fatty acid, eicosapentaenoic acid (EPA), a dietary nutrient that balances inflammation to restore homeostasis. RvE1 reduced leakage of H-FABP by up to 86%, which supports our hypothesis that inflammation as a mechanism of injury can be targeted for therapy. H-FABP as a blood biomarker was tested in 40 patients admitted to Boston Medical Center for respiratory distress, (20 patients with and 20 patients without COVID infection). High levels of H-FABP correlated with clinically diagnosed CVD, diabetes, and end-stage renal disease (ESRD) in both patient groups. The level of H-FABP indicates not only CVD damage but is a valuable measure for patients with increased inflammation disease comorbidities.
Collapse
Affiliation(s)
- A Zheng
- Boston University, United States of America
| | - N Huang
- Boston University School of Medicine, United States of America
| | - D Bean
- Boston University School of Medicine, United States of America
| | | | - Jude Deeney
- Boston University School of Medicine, United States of America
| | - M Sagar
- Boston Medical Center, United States of America
| | | |
Collapse
|
8
|
Fang J, Chen W, Hou P, Liu Z, Zuo M, Liu S, Feng C, Han Y, Li P, Shi Y, Shao C. NAD + metabolism-based immunoregulation and therapeutic potential. Cell Biosci 2023; 13:81. [PMID: 37165408 PMCID: PMC10171153 DOI: 10.1186/s13578-023-01031-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/14/2023] [Indexed: 05/12/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a critical metabolite that acts as a cofactor in energy metabolism, and serves as a cosubstrate for non-redox NAD+-dependent enzymes, including sirtuins, CD38 and poly(ADP-ribose) polymerases. NAD+ metabolism can regulate functionality attributes of innate and adaptive immune cells and contribute to inflammatory responses. Thus, the manipulation of NAD+ bioavailability can reshape the courses of immunological diseases. Here, we review the basics of NAD+ biochemistry and its roles in the immune response, and discuss current challenges and the future translational potential of NAD+ research in the development of therapeutics for inflammatory diseases, such as COVID-19.
Collapse
Affiliation(s)
- Jiankai Fang
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Wangwang Chen
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Pengbo Hou
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Zhanhong Liu
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Muqiu Zuo
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Shisong Liu
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Chao Feng
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Yuyi Han
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Peishan Li
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| | - Yufang Shi
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Changshun Shao
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
9
|
Consonni FM, Durante B, Manfredi M, Bleve A, Pandolfo C, Garlatti V, Vanella VV, Marengo E, Barberis E, Bottazzi B, Bombace S, My I, Condorelli G, Torri V, Sica A. Immunometabolic interference between cancer and COVID-19. Front Immunol 2023; 14:1168455. [PMID: 37063865 PMCID: PMC10090695 DOI: 10.3389/fimmu.2023.1168455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Even though cancer patients are generally considered more susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the mechanisms driving their predisposition to severe forms of coronavirus disease 2019 (COVID-19) have not yet been deciphered. Since metabolic disorders are associated with homeostatic frailty, which increases the risk of infection and cancer, we asked whether we could identify immunometabolic pathways intersecting with cancer and SARS-CoV-2 infection. Thanks to a combined flow cytometry and multiomics approach, here we show that the immunometabolic traits of COVID-19 cancer patients encompass alterations in the frequency and activation status of circulating myeloid and lymphoid subsets, and that these changes are associated with i) depletion of tryptophan and its related neuromediator tryptamine, ii) accumulation of immunosuppressive tryptophan metabolites (i.e., kynurenines), and iii) low nicotinamide adenine dinucleotide (NAD+) availability. This metabolic imbalance is accompanied by altered expression of inflammatory cytokines in peripheral blood mononuclear cells (PBMCs), with a distinctive downregulation of IL-6 and upregulation of IFNγ mRNA expression levels. Altogether, our findings indicate that cancer not only attenuates the inflammatory state in COVID-19 patients but also contributes to weakening their precarious metabolic state by interfering with NAD+-dependent immune homeostasis.
Collapse
Affiliation(s)
- Francesca Maria Consonni
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
| | - Barbara Durante
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Augusto Bleve
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
| | - Chiara Pandolfo
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Valentina Garlatti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
| | - Virginia Vita Vanella
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Barbara Bottazzi
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
| | - Sara Bombace
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Italy
| | - Ilaria My
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Italy
| | - Gianluigi Condorelli
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele-Milan, Italy
| | - Valter Torri
- Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- IRCCS Humanitas Clinical and Research Centre, Rozzano, Milan, Italy
- *Correspondence: Antonio Sica,
| |
Collapse
|
10
|
Carbone JT, Holzer KJ, Clift J, Fu Q. Latent profiles of biological dysregulation and risk of mortality: time-to-event analysis using the Midlife in the US longitudinal study. J Epidemiol Community Health 2023; 77:182-188. [PMID: 36627117 DOI: 10.1136/jech-2021-218073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND There is a well-established relationship between high allostatic load (AL) and increased risk of mortality. This study expands on the literature by combined latent profile analysis (LPA) with survival data analysis techniques to assess the degree to which AL status is associated with time to death. METHODS LPA was employed to identify underlying classes of biological dysregulation among a sample of 815 participants from the Midlife in the US study. Sex-stratified Cox proportional hazards regression models were used to estimate the association between class of biological dysregulation and time to death while controlling for sociodemographic covariates. RESULTS The LPA resulted in three classes: low dysregulation, immunometabolic dysregulation and parasympathetic reactivity. Women in the immunometabolic dysregulation group had more than three times the risk of death as compared with women in the low dysregulation group (HR=3.25, 95% CI: 1.47 to 7.07), but that there was not a statistically significant difference between the parasympathetic reactivity group and the low dysregulation group (HR=1.80, 95% CI: 0.62 to 5.23). For men, the risk of death for those in the immunometabolic dysregulation (HR=1.79, 95% CI: 0.88 to 3.65) and parasympathetic reactivity (HR=0.90, 95% CI: 0.34 to 3.65) groups did not differ from the low dysregulation group. CONCLUSION The findings are consistent with the previous research that demonstrates increased AL as a risk factor for mortality. Specifically, in women, that increased risk may be associated with immunometabolic dysregulation and not simply a generalised measure of cumulative risk as is typically employed in AL research.
Collapse
Affiliation(s)
- Jason T Carbone
- School of Social Work, Wayne State University, Detroit, Michigan, USA
| | - Katherine J Holzer
- St Louis School of Medicine, Washington University, St Louis, Missouri, USA
| | - Jennifer Clift
- School of Social Work, Wayne State University, Detroit, Michigan, USA
| | - Qiang Fu
- Department of Community Health, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
11
|
Cancer Patients and the COVID-19 Vaccines: Considerations and Challenges. Cancers (Basel) 2022; 14:cancers14225630. [PMID: 36428722 PMCID: PMC9688380 DOI: 10.3390/cancers14225630] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
Abstract
Few guidelines exist for COVID-19 vaccination amongst cancer patients, fostering uncertainty regarding the immunogenicity, safety, and effects of cancer therapies on vaccination, which this review aims to address. A literature review was conducted to include the latest articles covering the immunogenicity and safety of COVID-19 vaccination in patients with solid and hematologic cancers receiving various treatments. Lower seropositivity following vaccination was associated with malignancy (compared to the general population), and hematologic malignancy (compared to solid cancers). Patients receiving active cancer therapy (unspecified), chemotherapy, radiotherapy, and immunosuppressants generally demonstrated lower seropositivity compared to healthy controls; though checkpoint inhibition, endocrine therapy, and cyclin dependent kinase inhibition did not appear to affect seropositivity. Vaccination appeared safe and well-tolerated in patients with current or past cancer and those undergoing treatment. Adverse events were comparable to the general population, but inflammatory lymphadenopathy following vaccination was commonly reported and may be mistaken for malignant etiology. Additionally, radiation recall phenomenon was sporadically reported in patients who had received radiotherapy. Overall, while seropositivity rates were decreased, cancer patients showed capacity to generate safe and effective immune responses to COVID-19 vaccination, thus vaccination should be encouraged and hesitancy should be addressed in this population.
Collapse
|
12
|
Alaeddini M, Etemad-Moghadam S. SARS-Cov-2 infection in cancer patients, susceptibility, outcome and care. Am J Med Sci 2022; 364:511-520. [PMID: 35605680 PMCID: PMC9119956 DOI: 10.1016/j.amjms.2022.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 01/25/2023]
Abstract
The COVID-19 pandemic has led to many problems in cancer patients, which in part are due to insufficient knowledge of the exact implications of the virus on these individuals. Perceptions based on known facts about previous pandemics and coronaviruses might not agree with actual real-life experience and objective findings. We present a compilation of scientific facts and actual observations on different aspects of SARS-CoV-2 infection in cancer patients. These patients are at increased risk of viral contraction and have higher chances of severe disease/mortality. The latter is impacted by other factors and is still debated. In contrast to preliminary impressions, the benefits of anti-cancer treatments outweigh their risks and should be continued. Cancer patients generate antibodies in response to vaccination but in lower amounts than healthy people, especially those with hematologic cancers. Boosters, including third doses, have shown increased immune-responses in most patients. Vaccination should be prioritized in these individuals.
Collapse
Affiliation(s)
| | - Shahroo Etemad-Moghadam
- Corresponding author at: Shahroo Etemad-Moghadam, Dentistry Research Institute, Dental Research Centre, Ghods St, Enghelab Ave, P.O. Box: 14155-5583; Postal code: 14174, Tehran, Iran
| |
Collapse
|
13
|
Huang Y, Wang Y, Xu D, Xiao L, Qin W, Liu B, Yuan X. Characterization of the SARS-CoV-2 co-receptor NRP1 expression profiles in healthy people and cancer patients: Implication for susceptibility to COVID-19 disease and potential therapeutic strategy. Front Genet 2022; 13:995736. [PMID: 36338984 PMCID: PMC9627153 DOI: 10.3389/fgene.2022.995736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/10/2022] [Indexed: 08/03/2023] Open
Abstract
Neuropilin-1 (NRP1) is a transmembrane protein involved in many physiological and pathological processes, and it functions as a co-receptor to facilitate the entry of SARS-CoV-2 into host cells. Therefore, it is critical to predict the susceptibility to SARS-CoV-2 and prognosis after infection among healthy people and cancer patients based on expression of NRP1. In the current study, we analyzed the conservation and isoform of NRP1 using public databases. NRP1 expression landscape in healthy people, COVID-19 patients, and cancer patients at both bulk and single-cell RNA-seq level was also depicted. We also analyzed the relationship between tissue-specific NRP1 expression and overall survival (OS), as well as tumor immune environment at a pan-cancer level, providing a comprehensive insight into the relationship between the vulnerability to SARS-CoV-2 infection and tumorigenesis. In conclusion, we identified NRP1 as a potential biomarker in predicting susceptibility to SARS-CoV-2 infection among healthy people and cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Wan Qin
- *Correspondence: Wan Qin, ; Bo Liu, ; Xianglin Yuan,
| | - Bo Liu
- *Correspondence: Wan Qin, ; Bo Liu, ; Xianglin Yuan,
| | - Xianglin Yuan
- *Correspondence: Wan Qin, ; Bo Liu, ; Xianglin Yuan,
| |
Collapse
|
14
|
Identification of the effects of COVID-19 on patients with pulmonary fibrosis and lung cancer: a bioinformatics analysis and literature review. Sci Rep 2022; 12:16040. [PMID: 36163484 PMCID: PMC9512912 DOI: 10.1038/s41598-022-20040-x] [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: 06/19/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) poses a serious threat to human health and life. The effective prevention and treatment of COVID-19 complications have become crucial to saving patients’ lives. During the phase of mass spread of the epidemic, a large number of patients with pulmonary fibrosis and lung cancers were inevitably infected with the SARS-CoV-2 virus. Lung cancers have the highest tumor morbidity and mortality rates worldwide, and pulmonary fibrosis itself is one of the complications of COVID-19. Idiopathic lung fibrosis (IPF) and various lung cancers (primary and metastatic) become risk factors for complications of COVID-19 and significantly increase mortality in patients. Therefore, we applied bioinformatics and systems biology approaches to identify molecular biomarkers and common pathways in COVID-19, IPF, colorectal cancer (CRC) lung metastasis, SCLC and NSCLC. We identified 79 DEGs between COVID-19, IPF, CRC lung metastasis, SCLC and NSCLC. Meanwhile, based on the transcriptome features of DSigDB and common DEGs, we identified 10 drug candidates. In this study, 79 DEGs are the common core genes of the 5 diseases. The 10 drugs were found to have positive effects in treating COVID-19 and lung cancer, potentially reducing the risk of pulmonary fibrosis.
Collapse
|
15
|
Ahmad S, Matalon S, Kuebler WM. Understanding COVID-19 susceptibility and presentation based on its underlying physiology. Physiol Rev 2022; 102:1579-1585. [PMID: 35583178 PMCID: PMC9255705 DOI: 10.1152/physrev.00008.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Shama Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Sadis Matalon
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | | |
Collapse
|
16
|
Computational identification of host genomic biomarkers highlighting their functions, pathways and regulators that influence SARS-CoV-2 infections and drug repurposing. Sci Rep 2022; 12:4279. [PMID: 35277538 PMCID: PMC8915158 DOI: 10.1038/s41598-022-08073-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 02/15/2022] [Indexed: 12/13/2022] Open
Abstract
The pandemic threat of COVID-19 has severely destroyed human life as well as the economy around the world. Although, the vaccination has reduced the outspread, but people are still suffering due to the unstable RNA sequence patterns of SARS-CoV-2 which demands supplementary drugs. To explore novel drug target proteins, in this study, a transcriptomics RNA-Seq data generated from SARS-CoV-2 infection and control samples were analyzed. We identified 109 differentially expressed genes (DEGs) that were utilized to identify 10 hub-genes/proteins (TLR2, USP53, GUCY1A2, SNRPD2, NEDD9, IGF2, CXCL2, KLF6, PAG1 and ZFP36) by the protein–protein interaction (PPI) network analysis. The GO functional and KEGG pathway enrichment analyses of hub-DEGs revealed some important functions and signaling pathways that are significantly associated with SARS-CoV-2 infections. The interaction network analysis identified 5 TFs proteins and 6 miRNAs as the key regulators of hub-DEGs. Considering 10 hub-proteins and 5 key TFs-proteins as drug target receptors, we performed their docking analysis with the SARS-CoV-2 3CL protease-guided top listed 90 FDA approved drugs. We found Torin-2, Rapamycin, Radotinib, Ivermectin, Thiostrepton, Tacrolimus and Daclatasvir as the top ranked seven candidate drugs. We investigated their resistance performance against the already published COVID-19 causing top-ranked 11 independent and 8 protonated receptor proteins by molecular docking analysis and found their strong binding affinities, which indicates that the proposed drugs are effective against the state-of-the-arts alternatives independent receptor proteins also. Finally, we investigated the stability of top three drugs (Torin-2, Rapamycin and Radotinib) by using 100 ns MD-based MM-PBSA simulations with the two top-ranked proposed receptors (TLR2, USP53) and independent receptors (IRF7, STAT1), and observed their stable performance. Therefore, the proposed drugs might play a vital role for the treatment against different variants of SARS-CoV-2 infections.
Collapse
|
17
|
Qian W, Ye Y, Zuo L, Song T, Xu Q, Wang Y, Qian J, Tian Y. Immune checkpoint inhibitors use and effects on prognosis of COVID-19 infection: a systematic review and meta-analysis. Immunotherapy 2021; 13:1271-1282. [PMID: 34431319 PMCID: PMC8388614 DOI: 10.2217/imt-2021-0007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Aim: We aimed to quantify the effects of immune checkpoint inhibitors (ICIs) on the prognosis of COVID-19. Materials & methods: A meta-analysis was conducted and the hospitalization, severe disease and mortality rates were assessed. Thirteen studies comprising of 4614 cancer patients with COVID-19 were included. Results: When compared with cancer patients without prior ICI exposure, patients with prior ICI treatment exhibited a higher rate of hospitalization (odds ratio [OR] 2.0, 95% CI 1.19-3.38, p = 0.01). However, the OR of severe disease and mortality in ICI exposed cases was similar to non-ICI exposed patients (OR 1.55, 95% CI 0.69-3.51, p = 0.29; OR 1.12, 95% CI 0.85-1.48, p = 0.42, respectively). Conclusion: It is uncertain whether prior exposure to ICIs increases the risk of severe disease and death, however the observed OR suggest a higher rate of hospitalization.
Collapse
Affiliation(s)
- Wenwei Qian
- Department of General Surgery, Jinling Hospital, Medical School of Southeast University, No. 305 East Zhongshan Road, Nanjing, PR China
| | - Ying Ye
- Emergency Center, Affiliated Hospital of Xuzhou Medical College. 99 Huaihaixi Road, Xuzhou, Jiangsu, 221002, China
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, China
| | - Ting Song
- Department of Infectious Diseases, The Sixth People’s Hospital of Qingdao, No. 9 Fushun Road, Qingdao, Shandong, 266033, China
| | - Qing Xu
- Department of Oncology, Tongji University Cancer Center, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology & Nutrition, The University of MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jun Qian
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yun Tian
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| |
Collapse
|
18
|
78495111110.1152/physrev.00046.2020" />
Abstract
This medical review addresses the hypothesis that CD38/NADase is at the center of a functional axis (i.e., intracellular Ca2+ mobilization/IFNγ response/reactive oxygen species burst) driven by severe acute respiratory syndrome coronavirus 2 infection, as already verified in respiratory syncytial virus pathology and CD38 activity in other cellular settings. Key features of the hypothesis are that 1) the substrates of CD38 (e.g., NAD+ and NADP+) are depleted by viral-induced metabolic changes; 2) the products of the enzymatic activity of CD38 [e.g., cyclic adenosine diphosphate-ribose (ADPR)/ADPR/nicotinic acid adenine dinucleotide phosphate] and related enzymes [e.g., poly(ADP-ribose)polymerase, Sirtuins, and ADP-ribosyl hydrolase] are involved in the anti‐viral and proinflammatory response that favors the onset of lung immunopathology (e.g., cytokine storm and organ fibrosis); and 3) the pathological changes induced by this kinetic mechanism may be reduced by distinct modulators of the CD38/NAD+ axis (e.g., CD38 blockers, NAD+ suppliers, among others). This view is supported by arrays of associative basic and applied research data that are herein discussed and integrated with conclusions reported by others in the field of inflammatory, immune, tumor, and viral diseases.
Collapse
Affiliation(s)
- Alberto L. Horenstein
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Angelo C. Faini
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Fabio Malavasi
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| |
Collapse
|
19
|
Sinha S, Kundu CN. Cancer and COVID-19: Why are cancer patients more susceptible to COVID-19? Med Oncol 2021; 38:101. [PMID: 34302557 PMCID: PMC8302962 DOI: 10.1007/s12032-021-01553-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/20/2021] [Indexed: 12/20/2022]
Abstract
The COVID-19 pandemic has engulfed the entire world and has claimed more than 3 million lives worldwide. This viral disease is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and mainly characterized by fever, dry cough, fatigue, anosmia, anorexia, and dyspnea. The severity of the disease increases with age and presence of comorbidities, including cancer. Multiple clinical studies have shown that the cancer patients are highly susceptible to the severe form of the viral disease. In this review article, we have summarized the available scientific literature regarding the molecular links between COVID-19 and cancer, which make the cancer patients highly susceptible to COVID-19. Few studies have shown that the angiotensin-converting enzyme 2 (ACE2) receptor, transmembrane protease serine 2 (TMPRSS2), and the immune response and inflammation establish the interconnection between the two diseases. Additionally, we have also discussed whether SARS-CoV-2 can contribute to cancer development in COVID-19 patients. A recent study has suggested that SARS-CoV-2 may create a microenvironment that may support cancer cell proliferation and induce the activation of dormant cancer cells (DCCs). In another study, the blood sera of COVID-19 patients were found to activate epithelial-to-mesenchymal transition (EMT) in cancer cells. Overall, this review article will surely help the scientific community to understand why the cancer patients are so much prone to COVID-19 and will also motivate the researchers to find new therapeutic strategies that may save the lives of many COVID-19-infected cancer patients.
Collapse
Affiliation(s)
- Saptarshi Sinha
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, KIIT Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, KIIT Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
20
|
Obando-Pereda G. Can molecular mimicry explain the cytokine storm of SARS-CoV-2?: An in silico approach. J Med Virol 2021; 93:5350-5357. [PMID: 33913542 PMCID: PMC8242519 DOI: 10.1002/jmv.27040] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
PARP14 and PARP9 play a key role in macrophage immune regulation. SARS‐CoV‐2 is an emerging viral disease that triggers hyper‐inflammation known as a cytokine storm. In this study, using in silico tools, we hypothesize about the immunological phenomena of molecular mimicry between SARS‐CoV‐2 Nsp3 and the human PARP14 and PARP9. The results showed an epitope of SARS‐CoV‐2 Nsp3 protein that contains consensus sequences for both human PARP14 and PARP9 that are antigens for MHC Classes 1 and 2, which can potentially induce an immune response against human PARP14 and PARP9; while its depletion causes a hyper‐inflammatory state in SARS‐CoV‐2 patients. Molecular mimicry could produce an inflammatory state in SARS‐CoV‐2 patients. Human PARP14 and PARP9 are the proteins involves in this phenomena.
Collapse
Affiliation(s)
- Gustavo Obando-Pereda
- Proteomics, Inflammation and Pain Laboratory, Universidad Católica de Santa María, Arequipa, Peru
| |
Collapse
|
21
|
Abstract
This medical review addresses the hypothesis that CD38/NADase is at the center of a functional axis (i.e., intracellular Ca2+ mobilization/IFNγ response/reactive oxygen species burst) driven by severe acute respiratory syndrome coronavirus 2 infection, as already verified in respiratory syncytial virus pathology and CD38 activity in other cellular settings. Key features of the hypothesis are that 1) the substrates of CD38 (e.g., NAD+ and NADP+) are depleted by viral-induced metabolic changes; 2) the products of the enzymatic activity of CD38 [e.g., cyclic adenosine diphosphate-ribose (ADPR)/ADPR/nicotinic acid adenine dinucleotide phosphate] and related enzymes [e.g., poly(ADP-ribose)polymerase, Sirtuins, and ADP-ribosyl hydrolase] are involved in the anti‐viral and proinflammatory response that favors the onset of lung immunopathology (e.g., cytokine storm and organ fibrosis); and 3) the pathological changes induced by this kinetic mechanism may be reduced by distinct modulators of the CD38/NAD+ axis (e.g., CD38 blockers, NAD+ suppliers, among others). This view is supported by arrays of associative basic and applied research data that are herein discussed and integrated with conclusions reported by others in the field of inflammatory, immune, tumor, and viral diseases.
Collapse
Affiliation(s)
- Alberto L Horenstein
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Angelo C Faini
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Fabio Malavasi
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| |
Collapse
|
22
|
Xiao N, Nie M, Pang H, Wang B, Hu J, Meng X, Li K, Ran X, Long Q, Deng H, Chen N, Li S, Tang N, Huang A, Hu Z. Integrated cytokine and metabolite analysis reveals immunometabolic reprogramming in COVID-19 patients with therapeutic implications. Nat Commun 2021; 12:1618. [PMID: 33712622 PMCID: PMC7955129 DOI: 10.1038/s41467-021-21907-9] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/19/2021] [Indexed: 01/08/2023] Open
Abstract
Cytokine release syndrome (CRS) is a major cause of the multi-organ injury and fatal outcome induced by SARS-CoV-2 infection in severe COVID-19 patients. Metabolism can modulate the immune responses against infectious diseases, yet our understanding remains limited on how host metabolism correlates with inflammatory responses and affects cytokine release in COVID-19 patients. Here we perform both metabolomics and cytokine/chemokine profiling on serum samples from healthy controls, mild and severe COVID-19 patients, and delineate their global metabolic and immune response landscape. Correlation analyses show tight associations between metabolites and proinflammatory cytokines/chemokines, such as IL-6, M-CSF, IL-1α, IL-1β, and imply a potential regulatory crosstalk between arginine, tryptophan, purine metabolism and hyperinflammation. Importantly, we also demonstrate that targeting metabolism markedly modulates the proinflammatory cytokines release by peripheral blood mononuclear cells isolated from SARS-CoV-2-infected rhesus macaques ex vivo, hinting that exploiting metabolic alterations may be a potential strategy for treating fatal CRS in COVID-19. Metabolism changes can modulate immune responses in many contexts, and vice versa. Here the authors associate metabolomic, as well as cytokine and chemokine, data from stratified COVID-19 patients to find that arginine, tryptophan and purine metabolic pathways correlate with hyperproliferation, thus hinting at potential therapeutic targets for severe COVID-19 patients.
Collapse
Affiliation(s)
- Nan Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Meng Nie
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Huanhuan Pang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bohong Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jieli Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Xiangjun Meng
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Ke Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xiaorong Ran
- Agilent Technologies (China), Chaoyang District, Beijing, 100102, China
| | - Quanxin Long
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Haijun Deng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Na Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Shao Li
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China.
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China.
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China. .,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China. .,Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
23
|
Trama A, Proto C, Whisenant JG, Torri V, Cortellini A, Michielin O, Barlesi F, Dingemans AMC, Van Meerbeeck J, Pancaldi V, Mazieres J, Soo RA, Leighl NB, Peters S, Wakelee H, Horn L, Hellmann M, Wong SK, Garassino MC, Baena J. Supporting Clinical Decision-Making during the SARS-CoV-2 Pandemic through a Global Research Commitment: The TERAVOLT Experience. Cancer Cell 2020; 38:602-604. [PMID: 33091381 PMCID: PMC7534826 DOI: 10.1016/j.ccell.2020.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To understand the real impact of COVID-19 on cancer patients, an entirely new data collection effort was initiated within the Thoracic Cancers International COVID-19 Collaboration (TERAVOLT). TERAVOLT reported high mortality related to COVID-19 infection in thoracic cancer patients and identified several negative prognostic factors. In this commentary, we discuss the importance and limits of patient registries to support decision-making in thoracic cancer during the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Annalisa Trama
- Department of Research, Evaluative Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Proto
- Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Jennifer G Whisenant
- Department of Medicine, Division of Hematology & Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Valter Torri
- Laboratory of Clinical Research, Methodology Oncology Department "Mario Negri" Institute of Pharmacological Researches, IRCCS, Milan, Italy
| | - Alessio Cortellini
- Department of Biotechnology and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | - Olivier Michielin
- Oncology Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Fabrice Barlesi
- Gustave Roussy Cancer Center, Villejuif, France; Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France
| | - Anne-Marie C Dingemans
- Department of Pulmonology, Erasmus University Medical Center, Rotterdam, and University Maastricht, Maastricht, the Netherlands
| | - Jan Van Meerbeeck
- Department of Pulmonology and Thoracic Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Vera Pancaldi
- University Paul Sabatier, Toulouse, Inserm, Centre de Recherche en Cancérologie de Toulouse, Toulouse, France; Barcelona Supercomputing Center, Barcelona
| | - Julien Mazieres
- Institut Universitaire du Cancer, Centre Hospitalier Universitaire, Toulouse, France
| | - Ross A Soo
- National University Cancer Institute, Singapore
| | | | - Solange Peters
- Oncology Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Heather Wakelee
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, USA
| | - Leora Horn
- Department of Medicine, Division of Hematology & Oncology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew Hellmann
- Medical Oncology, Memorial Sloan Kettering Evelyn H. Lauder Breast Center, New York, NY, USA
| | - Selina K Wong
- Department of Medicine, Division of Hematology & Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marina Chiara Garassino
- Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Javier Baena
- Medical Oncology Department, Thoracic Cancer and Early Drug Development Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
24
|
Sriram K, Insel PA. Inflammation and thrombosis in COVID-19 pathophysiology: proteinase-activated and purinergic receptors as drivers and candidate therapeutic targets. Physiol Rev 2020; 101:545-567. [PMID: 33124941 PMCID: PMC8238137 DOI: 10.1152/physrev.00035.2020] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Evolving information has identified disease mechanisms and dysregulation of host biology that might be targeted therapeutically in coronavirus disease 2019 (COVID-19). Thrombosis and coagulopathy, associated with pulmonary injury and inflammation, are emerging clinical features of COVID-19. We present a framework for mechanisms of thrombosis in COVID-19 that initially derive from interaction of SARS-CoV-2 with ACE2, resulting in dysregulation of angiotensin signaling and subsequent inflammation and tissue injury. These responses result in increased signaling by thrombin (proteinase-activated) and purinergic receptors, which promote platelet activation and exert pathological effects on other cell types (e.g., endothelial cells, epithelial cells, and fibroblasts), further enhancing inflammation and injury. Inhibitors of thrombin and purinergic receptors may, thus, have therapeutic effects by blunting platelet-mediated thromboinflammation and dysfunction in other cell types. Such inhibitors include agents (e.g., anti-platelet drugs) approved for other indications, and that could be repurposed to treat, and potentially improve the outcome of, COVID-19 patients. COVID-19, caused by the SARS-CoV-2 virus, drives dysregulation of angiotensin signaling, which, in turn, increases thrombin-mediated and purinergic-mediated activation of platelets and increase in inflammation. This thromboinflammation impacts the lungs and can also have systemic effects. Inhibitors of receptors that drive platelet activation or inhibitors of the coagulation cascade provide opportunities to treat COVID-19 thromboinflammation.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Paul A Insel
- Department of Pharmacology and Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|