1
|
Al-Naimi MS, Abu-Raghif AR, Mansoor AFA, Fawzi HA. Isofraxidin Attenuates Lipopolysaccharide-Induced Cytokine Release in Mice Lung and Liver Tissues via Inhibiting Inflammation and Oxidative Stress. Biomedicines 2025; 13:653. [PMID: 40149629 PMCID: PMC11940160 DOI: 10.3390/biomedicines13030653] [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: 01/22/2025] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Isofraxidin is a hydroxylcoumarin derived from herbal Fraxinus and Eleutherococcus. It has been shown that isofraxidin has antioxidant, anti-inflammatory, anti-diabetic, and anti-lipidemic effects. The study aimed to examine the therapeutic effects of isofraxidin with and without methylprednisolone to ameliorate lipopolysaccharide (LPS)-induced cytokine-releasing syndrome. Methods: The study comprised two phases: preventive and therapeutic. In all the experiments that involved LPS induction, a single dose of LPS (5 mg/kg) was used. The preventive phase involved the administration of the agents before LPS induction, in which 50 mg/kg of methylprednisolone, 15 mg/kg of isofraxidin, or a combination of 7.5 mg/kg of isofraxidin plus 25 mg/kg methylprednisolone were given daily for 3 days before induction. The therapeutic phase involved the administration of the following agents after LPS induction: 50 mg/kg methylprednisolone, 15 mg/kg of isofraxidin, or a combination of 7.5 mg/kg of isofraxidin plus 25 mg/kg methylprednisolone were given once daily was given for 7 days. Results: Isofraxidin treatment with or without methylprednisolone ameliorates LPS-induced inflammatory and oxidative stress damage in mice; it reduces the inflammatory (IL-6, TNF-α, IL-1β, IL-8, Malondialdehyde, and IFN-γ) and oxidative stress markers. Additionally, isofraxidin treatment with or without methylprednisolone prevented liver and lung tissue damage induced by LPS. Conclusions: Isofraxidin exhibited preventive and therapeutic properties against lipopolysaccharide-induced cytokine storms in mice via anti-inflammatory and antioxidant pathways, and its combination with methylprednisolone demonstrated synergistic outcomes.
Collapse
Affiliation(s)
- Marwa Salih Al-Naimi
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad 10006, Iraq;
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Farahidi University, Baghdad 00965, Iraq
| | - Ahmed R. Abu-Raghif
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad 10006, Iraq;
| | - Ahmed F. Abed Mansoor
- Department of Pharmacology and Toxicology, College of Pharmacy, National University of Science and Technology, Nasiriyah 64001, Iraq;
| | - Hayder Adnan Fawzi
- Department of Clinical Pharmacy, College of Pharmacy, AlMustafa University, Baghdad 10064, Iraq;
| |
Collapse
|
2
|
Sun W, Wang S, Han J, Zhuo L, Cao J, Zhou F. Symptoms of Hematologic Tumors Patients after CAR-T Therapy: A Systematic Review and Meta-Analysis. J Pain Symptom Manage 2025; 69:304-317. [PMID: 39547263 DOI: 10.1016/j.jpainsymman.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/28/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
CONTEXT Patients with hematologic neoplasms after Chimeric antigen receptor T-cell (CAR-T) therapy have multiple syndromes, with corresponding symptoms. OBJECTIVES The review aimed to integrate the severity and incidences of symptoms in these patients, and to investigate the difference of the symptoms among different geographic locations, types of hematological tumors, evaluating instruments, and evaluating time, to provide a theoretical basis for symptom management. METHODS A literature search of PubMed, Web of Science, Embase, the Cochrane Library, China National Knowledge Internet, SinoMed, VIP, and WANFANG DATA was performed for studies reporting symptom scores or symptom incidences of these patients published before November 9, 2023. Heterogeneity between studies was assessed by Higgins' I2. A random effects model was used for studies with I2 > 50%. Methodological quality of included studies was assessed using the Joanna Briggs Institute Critical Appraisal Checklist. RESULTS Eight studies were included. Among the reported symptoms, sleep disturbance, fatigue and depression were of higher severity, with the standardized scores exceeding 50. Sadness, problem with concentration, problem with memory, cough and nausea were the top five symptoms in incidence, which exceeded 50%. The symptom scores and incidences assessed by the patient-reported outcomes were higher. Within 90 days of CAR-T infusion, these patients reported a significantly higher severity and incidence of multiple symptoms. CONCLUSION Patients with hematologic neoplasms treated by CAR-T suffer from multiple symptoms, including depression, fatigue, and so on. Instruments used to evaluate symptoms and the evaluating time may influence the outcome of symptom assessment.
Collapse
Affiliation(s)
- Wan Sun
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Shuo Wang
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Jiachen Han
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Lang Zhuo
- School of Public Health (L.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Jiang Cao
- Department of Hematology (J.C.), The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Fang Zhou
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China.
| |
Collapse
|
3
|
Sawant-Basak A, Olabode D, Dai D, Vishwanathan K, Phipps A. Assessing Trends in Cytokine-CYP Drug Interactions and Relevance to Drug Dosing. Drug Metab Dispos 2024; 52:1196-1200. [PMID: 38383116 DOI: 10.1124/dmd.123.001499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/12/2024] [Accepted: 02/08/2024] [Indexed: 02/23/2024] Open
Abstract
The regulation of drug-metabolizing enzymes and transporters by cytokines has been extensively studied in vitro and in clinic. Cytokine-mediated suppression of cytochrome P450 (CYP) or drug transporters may increase or decrease the systemic clearance of drug substrates that are primarily cleared via these pathways; neutralization of cytokines by therapeutic proteins may thereby alter systemic exposures of such drug substrates. The Food and Drug Administration recommends evaluating such clinical drug interactions during clinical development and has provided labeling recommendations for therapeutic proteins. To determine the clinical relevance of these drug interactions to dose adjustments, trends in steady-state exposures of CYP-sensitive substrates coadministered with cytokine modulators as reported in the University of Washington Drug Interaction Database were extracted and examined for each of the CYPs. Coadministration of cytochrome P450 family 3 subfamily A (CYP3A) (midazolam/simvastatin), cytochrome P450 subfamily 2C19 (omeprazole), or cytochrome P450 subfamily 1A2 (caffeine/tizanidine) substrates with anti-interleukin-6 and with anti-interleukin-23 therapeutics led to changes in systemic exposures of CYP substrates ranging from ∼ -58% to ∼35%; no significant trends were observed for cytochrome P450 subfamily 2D6 (dextromethorphan) and cytochrome P450 subfamily 2C9 (warfarin) substrates. Although none of these changes in systemic exposures have been reported as clinically meaningful, dose adjustment of midazolam for optimal sedation in acute care settings has been reported. Simulated concentration-time profiles of midazolam under conditions of elevated cytokine levels when coadministered with tocilizumab, suggest a ∼six- to sevenfold increase in midazolam clearance, suggesting potential implications of cytokine-CYP drug interactions on dose adjustments of sensitive CYP3A substrates in acute care settings. Additionally, this article also provides a brief overview of nonclinical and clinical assessments of cytokine-CYP drug interactions in drug discovery and development. SIGNIFICANCE STATEMENT: There has been significant progress in understanding cytokine-mediated drug interactions for CYP-sensitive substrates. This article provides an overview of the progress in this field, including a trend analysis of systemic exposures of CYP-sensitive substrates coadministered with anti-interleukin therapeutics. In addition, the review also provides a perspective of current methods used to assess these drug interactions during drug development and a focus on individualized medicine, particularly in acute care settings.
Collapse
Affiliation(s)
- Aarti Sawant-Basak
- Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Waltham, Massachusetts (A.S.-B., D.O., K.V.); Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Gaithersburg, Massachusetts (D.D.); and Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Cambridge, UK (A.P.)
| | - Damilola Olabode
- Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Waltham, Massachusetts (A.S.-B., D.O., K.V.); Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Gaithersburg, Massachusetts (D.D.); and Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Cambridge, UK (A.P.)
| | - David Dai
- Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Waltham, Massachusetts (A.S.-B., D.O., K.V.); Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Gaithersburg, Massachusetts (D.D.); and Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Cambridge, UK (A.P.)
| | - Karthick Vishwanathan
- Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Waltham, Massachusetts (A.S.-B., D.O., K.V.); Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Gaithersburg, Massachusetts (D.D.); and Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Cambridge, UK (A.P.)
| | - Alex Phipps
- Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Waltham, Massachusetts (A.S.-B., D.O., K.V.); Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Gaithersburg, Massachusetts (D.D.); and Clinical Pharmacology and Safety Sciences, Oncology Research and Development AstraZeneca, Cambridge, UK (A.P.)
| |
Collapse
|
4
|
Sani F, Shojaei S, Tabatabaei SA, Khorraminejad-Shirazi M, Latifi M, Sani M, Azarpira N. CAR-T cell-derived exosomes: a new perspective for cancer therapy. Stem Cell Res Ther 2024; 15:174. [PMID: 38886844 PMCID: PMC11184895 DOI: 10.1186/s13287-024-03783-4] [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: 02/07/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell adoptive immunotherapy is a promising cancer treatment that uses genetically engineered T cells to attack tumors. However, this therapy can have some adverse effects. CAR-T cell-derived exosomes are a potential alternative to CAR-T cells that may overcome some limitations. Exosomes are small vesicles released by cells and can carry a variety of molecules, including proteins, RNA, and DNA. They play an important role in intercellular communication and can be used to deliver therapeutic agents to cancer cells. The application of CAR-T cell-derived exosomes could make CAR-T cell therapy more clinically controllable and effective. Exosomes are cell-free, which means that they are less likely to cause adverse reactions than CAR-T cells. The combination of CAR-T cells and exosomes may be a more effective way to treat cancer than either therapy alone. Exosomes can deliver therapeutic agents to cancer cells where CAR-T cells cannot reach. The appropriate application of both cellular and exosomal platforms could make CAR-T cell therapy a more practicable treatment for cancer. This combination therapy could offer a safe and effective way to treat a variety of cancers.
Collapse
Affiliation(s)
- Farnaz Sani
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shabnam Shojaei
- School of Medicine, Shiraz Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammadhossein Khorraminejad-Shirazi
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pathology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mona Latifi
- Department of Physiological Science, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Mahsa Sani
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Negar Azarpira
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Transplant Research Center, Shiraz University of Medical Sciences, Khalili Street, P.O. Box: 7193711351, Shiraz, Iran.
| |
Collapse
|
5
|
Zhou X, Geyer FK, Happel D, Takimoto J, Kolmar H, Rabinovich B. Using protein geometry to optimize cytotoxicity and the cytokine window of a ROR1 specific T cell engager. Front Immunol 2024; 15:1323049. [PMID: 38455046 PMCID: PMC10917902 DOI: 10.3389/fimmu.2024.1323049] [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: 10/17/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
T cell engaging bispecific antibodies have shown clinical proof of concept for hematologic malignancies. Still, cytokine release syndrome, neurotoxicity, and on-target-off-tumor toxicity, especially in the solid tumor setting, represent major obstacles. Second generation TCEs have been described that decouple cytotoxicity from cytokine release by reducing the apparent binding affinity for CD3 and/or the TAA but the results of such engineering have generally led only to reduced maximum induction of cytokine release and often at the expense of maximum cytotoxicity. Using ROR1 as our model TAA and highly modular camelid nanobodies, we describe the engineering of a next generation decoupled TCE that incorporates a "cytokine window" defined as a dose range in which maximal killing is reached but cytokine release may be modulated from very low for safety to nearly that induced by first generation TCEs. This latter attribute supports pro-inflammatory anti-tumor activity including bystander killing and can potentially be used by clinicians to safely titrate patient dose to that which mediates maximum efficacy that is postulated as greater than that possible using standard second generation approaches. We used a combined method of optimizing TCE mediated synaptic distance and apparent affinity tuning of the TAA binding arms to generate a relatively long but persistent synapse that supports a wide cytokine window, potent killing and a reduced propensity towards immune exhaustion. Importantly, this next generation TCE induced significant tumor growth inhibition in vivo but unlike a first-generation non-decoupled benchmark TCE that induced lethal CRS, no signs of adverse events were observed.
Collapse
Affiliation(s)
- Xueyuan Zhou
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| | - Felix Klaus Geyer
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Dominic Happel
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jeffrey Takimoto
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Brian Rabinovich
- Drug Discovery and Development, Fuse Biotherapeutics, Woburn, MA, United States
| |
Collapse
|
6
|
Krenytska D, Strubchevska K, Kozyk M, Vovk T, Halenova T, Kot L, Raksha N, Savchuk O, Falalyeyeva T, Tsyryuk O, Ostapchenko L. Circulating levels of inflammatory cytokines and angiogenesis-related growth factors in patients with osteoarthritis after COVID-19. Front Med (Lausanne) 2023; 10:1168487. [PMID: 37484856 PMCID: PMC10358362 DOI: 10.3389/fmed.2023.1168487] [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: 02/17/2023] [Accepted: 05/25/2023] [Indexed: 07/25/2023] Open
Abstract
Background The disease COVID-19, caused by SARS-CoV-2 infection, has a systemic effect and is associated with a number of pathophysiological mechanisms that mobilize a wide range of biomolecules. Cytokines and growth factors (GFs) are critical regulators of tissue damage or repair in osteoarthritis (OA) and are being recognized as key players in the pathogenesis of COVID-19. A clear understanding of the long-term consequences of SARS-CoV-2 infection, especially in patients with concomitant chronic diseases, is limited and needs to be elucidated. The study aimed to evaluate the degree of inflammation and levels of pro-angiogenic and hypoxic factors, as well as heat shock proteins HSP60 and HSP70 in plasma, of patients with OA after recovery from COVID-19. Methods The research involved patients of an orthopedic specialty clinic aged 39 to 80 diagnosed with knee OA. All examined patients were divided into three groups: the Control group included conditionally healthy donors, group OA included patients with knee OA mainly stage II or III and the group of OA and COVID-19 included patients with OA who had COVID-19. The plasma levels of pro-inflammatory molecules IL-1β, IL-6, TNF-α, NF-κB, angiogenic factors VEGF, FGF-2, PDGF, hypoxic factor HIF-1α and molecular chaperones HSP60 and HSP70 were measured by enzyme-linked immunosorbent assay. Results The study showed that in both groups of patients, with OA and convalescent COVID-19, there was an increase in the plasma level of IL-1β and a decrease in TNF-α and NF-κB levels when compared to healthy controls. Systemic deregulation of the cytokine profile was accompanied by reduction in plasma levels of pro-angiogenic growth factors, most pronounced in cases of VEGF and PDGF. This analysis did not reveal any significant difference in the plasma level of HIF-1α. A decrease in the level of stress protein HSP60 in the blood of patients with OA, as well as those patients who have had SARS-CoV-2 infection, has been established. Conclusion The results suggest the potential role pro-inflammatory cytokines and angiogenesis-related growth factors in pathogenesis of both joint pathologies and long-term systemic post-COVID-19 disorders.
Collapse
Affiliation(s)
- Daryna Krenytska
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | | - Marko Kozyk
- William Beaumont Hospital, Royal Oak, MI, United States
| | - Tetiana Vovk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetiana Halenova
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Larysa Kot
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Nataliia Raksha
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olexii Savchuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olena Tsyryuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Liudmyla Ostapchenko
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| |
Collapse
|
7
|
Shah D, Soper B, Shopland L. Cytokine release syndrome and cancer immunotherapies - historical challenges and promising futures. Front Immunol 2023; 14:1190379. [PMID: 37304291 PMCID: PMC10248525 DOI: 10.3389/fimmu.2023.1190379] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Cancer is the leading cause of death worldwide. Cancer immunotherapy involves reinvigorating the patient's own immune system to fight against cancer. While novel approaches like Chimeric Antigen Receptor (CAR) T cells, bispecific T cell engagers, and immune checkpoint inhibitors have shown promising efficacy, Cytokine Release Syndrome (CRS) is a serious adverse effect and remains a major concern. CRS is a phenomenon of immune hyperactivation that results in excessive cytokine secretion, and if left unchecked, it may lead to multi-organ failure and death. Here we review the pathophysiology of CRS, its occurrence and management in the context of cancer immunotherapy, and the screening approaches that can be used to assess CRS and de-risk drug discovery earlier in the clinical setting with more predictive pre-clinical data. Furthermore, the review also sheds light on the potential immunotherapeutic approaches that can be used to overcome CRS associated with T cell activation.
Collapse
Affiliation(s)
- Deep Shah
- In vivo Services, The Jackson Laboratory, Sacramento, CA, United States
| | - Brian Soper
- Technical Information Services, The Jackson Laboratory, Bar Harbor, ME, United States
| | - Lindsay Shopland
- In vivo Services, The Jackson Laboratory, Sacramento, CA, United States
| |
Collapse
|
8
|
Muacevic A, Adler JR, Nigh G, McCullough PA. A Potential Role of the Spike Protein in Neurodegenerative Diseases: A Narrative Review. Cureus 2023; 15:e34872. [PMID: 36788995 PMCID: PMC9922164 DOI: 10.7759/cureus.34872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 02/13/2023] Open
Abstract
Human prion protein and prion-like protein misfolding are widely recognized as playing a causal role in many neurodegenerative diseases. Based on in vitro and in vivo experimental evidence relating to prion and prion-like disease, we extrapolate from the compelling evidence that the spike glycoprotein of SARS-CoV-2 contains extended amino acid sequences characteristic of a prion-like protein to infer its potential to cause neurodegenerative disease. We propose that vaccine-induced spike protein synthesis can facilitate the accumulation of toxic prion-like fibrils in neurons. We outline various pathways through which these proteins could be expected to distribute throughout the body. We review both cellular pathologies and the expression of disease that could become more frequent in those who have undergone mRNA vaccination. Specifically, we describe the spike protein's contributions, via its prion-like properties, to neuroinflammation and neurodegenerative diseases; to clotting disorders within the vasculature; to further disease risk due to suppressed prion protein regulation in the context of widely prevalent insulin resistance; and to other health complications. We explain why these prion-like characteristics are more relevant to vaccine-related mRNA-induced spike proteins than natural infection with SARS-CoV-2. We note with an optimism an apparent loss of prion-like properties among the current Omicron variants. We acknowledge that the chain of pathological events described throughout this paper is only hypothetical and not yet verified. We also acknowledge that the evidence we usher in, while grounded in the research literature, is currently largely circumstantial, not direct. Finally, we describe the implications of our findings for the general public, and we briefly discuss public health recommendations we feel need urgent consideration. An earlier version of this article was previously posted to the Authorea preprint server on August 16, 2022.
Collapse
|
9
|
Wang SJ, Brodie KC, De Pons JL, Demos WM, Gibson AC, Hayman GT, Hill ML, Kaldunski ML, Lamers L, Laulederkind SJF, Nalabolu HS, Thota J, Thorat K, Tutaj MA, Tutaj M, Vedi M, Zacher S, Smith JR, Dwinell MR, Kwitek AE. Ontological Analysis of Coronavirus Associated Human Genes at the COVID-19 Disease Portal. Genes (Basel) 2022; 13:genes13122304. [PMID: 36553571 PMCID: PMC9777590 DOI: 10.3390/genes13122304] [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: 11/09/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic stemmed a parallel upsurge in the scientific literature about SARS-CoV-2 infection and its health burden. The Rat Genome Database (RGD) created a COVID-19 Disease Portal to leverage information from the scientific literature. In the COVID-19 Portal, gene-disease associations are established by manual curation of PubMed literature. The portal contains data for nine ontologies related to COVID-19, an embedded enrichment analysis tool, as well as links to a toolkit. Using these information and tools, we performed analyses on the curated COVID-19 disease genes. As expected, Disease Ontology enrichment analysis showed that the COVID-19 gene set is highly enriched with coronavirus infectious disease and related diseases. However, other less related diseases were also highly enriched, such as liver and rheumatic diseases. Using the comparison heatmap tool, we found nearly 60 percent of the COVID-19 genes were associated with nervous system disease and 40 percent were associated with gastrointestinal disease. Our analysis confirms the role of the immune system in COVID-19 pathogenesis as shown by substantial enrichment of immune system related Gene Ontology terms. The information in RGD's COVID-19 disease portal can generate new hypotheses to potentiate novel therapies and prevention of acute and long-term complications of COVID-19.
Collapse
Affiliation(s)
- Shur-Jen Wang
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kent C. Brodie
- Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jeffrey L. De Pons
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wendy M. Demos
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Adam C. Gibson
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - G. Thomas Hayman
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Morgan L. Hill
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary L. Kaldunski
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Logan Lamers
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stanley J. F. Laulederkind
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Harika S. Nalabolu
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jyothi Thota
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ketaki Thorat
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Marek A. Tutaj
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monika Tutaj
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mahima Vedi
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stacy Zacher
- Finance and Administration, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jennifer R. Smith
- The Rat Genome Database, Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Melinda R. Dwinell
- The Rat Genome Database, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anne E. Kwitek
- The Rat Genome Database, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence:
| |
Collapse
|
10
|
Fanelli M, Petrone V, Buonifacio M, Delibato E, Balestrieri E, Grelli S, Minutolo A, Matteucci C. Multidistrict Host-Pathogen Interaction during COVID-19 and the Development Post-Infection Chronic Inflammation. Pathogens 2022; 11:1198. [PMID: 36297256 PMCID: PMC9607297 DOI: 10.3390/pathogens11101198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 12/15/2022] Open
Abstract
Due to the presence of the ACE2 receptor in different tissues (nasopharynx, lung, nervous tissue, intestine, liver), the COVID-19 disease involves several organs in our bodies. SARS-CoV-2 is able to infect different cell types, spreading to different districts. In the host, an uncontrolled and altered immunological response is triggered, leading to cytokine storm, lymphopenia, and cellular exhaustion. Hence, respiratory distress syndrome (ARDS) and systemic multi-organ dysfunction syndrome (MODS) are established. This scenario is also reflected in the composition of the microbiota, the balance of which is regulated by the interaction with the immune system. A change in microbial diversity has been demonstrated in COVID-19 patients compared with healthy donors, with an increase in potentially pathogenic microbial genera. In addition to other symptoms, particularly neurological, the occurrence of dysbiosis persists after the SARS-CoV-2 infection, characterizing the post-acute COVID syndrome. This review will describe and contextualize the role of the immune system in unbalance and dysbiosis during SARS-CoV-2 infection, from the acute phase to the post-COVID-19 phase. Considering the tight relationship between the immune system and the gut-brain axis, the analysis of new, multidistrict parameters should be aimed at understanding and addressing chronic multisystem dysfunction related to COVID-19.
Collapse
Affiliation(s)
- Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Margherita Buonifacio
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Elisabetta Delibato
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Virology Unit, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
11
|
Borodin S, Tuharov Y, Ostapchenko D, Kot L, Korotkyi O. VIEW OF THE PATHOGENETIC MECHANISMS OF JOINT DAMAGE IN CORONAVIRUS DISEASE 2019. BULLETIN OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV. SERIES: BIOLOGY 2022. [DOI: 10.17721/1728.2748.2022.89.5-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The Сoronavirus disease 2019 (COVID-19) pandemic has had an extremely serious impact on the livelihoods of people worldwide. Despite the mainly respiratory manifestations of SARS-CoV-2 infection, its consequences can affect the functioning of most systems of organism, including the musculoskeletal, dysfunction of which is a leading factor in disability of the population. Diseases of the joints are one of the most common pathologie of modernity, which in the last decade are increasingly found in young people and even children. The musculoskeletal symptoms can be observed as isolated clinical signs and expressed regardless of the severity of the viral disease. Chronic fatigue, myalgia, swelling and joint pain may occur during the acute phase of COVID-19 and as short-term or long-term complications, but their prevalence has not been systematically studied. Considering the wide range of clinical manifestations of SARS-CoV-2 infection and the complexity of their pathogenesis, the mechanisms underlying lesions of musculoskeletal system and rheumatological complications remain unclear. Today, the main research in this direction is focused mainly on a complete understanding of the regulatory pathways of immune dysregulation and inflammation. Excessive secretion of pro-inflammatory cytokines, disruption of signal transduction and immune response are the result of the severe impact of SARS-CoV-2 infection on most organs, including joints, as well as the use of corticosteroids for the treatment of patients with COVID-19. The presented review highlights generalized information on the main pathophysiological processes that can occur in the joints as a manifestation of the impact of SARS-CoV-2 infection, and possible key mechanisms which contribute to the progression of pathological changes.
Collapse
|