1
|
Cappelletti G, Brambilla L, Strizzi S, Limanaqi F, Melzi V, Rizzuti M, Nizzardo M, Saulle I, Trabattoni D, Corti S, Clerici M, Biasin M. iPSC-derived human cortical organoids display profound alterations of cellular homeostasis following SARS-CoV-2 infection and Spike protein exposure. FASEB J 2025; 39:e70396. [PMID: 39950320 PMCID: PMC11826378 DOI: 10.1096/fj.202401604rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025]
Abstract
COVID-19 commonly leads to respiratory issues, yet numerous patients also exhibit a diverse range of neurological conditions, suggesting a detrimental impact of SARS-CoV-2 or the viral Spike protein on the central nervous system. Nonetheless, the molecular pathway behind neurological pathology and the presumed neurotropism of SARS-CoV-2 remains largely unexplored. We generated human cortical organoids (HCOs) derived from human induced pluripotent stem cells (hiPSC) to assess: (1) the expression of SARS-CoV-2 main entry factors; (2) their vulnerability to SARS-CoV-2 infection; and (3) the impact of SARS-CoV-2 infection and exposure to the Spike protein on their transcriptome. Results proved that (1) HCOs express the main SARS-CoV-2 receptors and co-receptors; (2) HCOs may be productively infected by SARS-CoV-2; (3) the viral particles released by SARS-CoV-2-infected HCOs are able to re-infect another cellular line; and (4) the infection resulted in the activation of apoptotic and stress pathways, along with inflammatory processes. Notably, these effects were recapitulated when HCOs were exposed to the Spike protein alone. The data obtained demonstrate that SARS-CoV-2 likely infects HCOs probably through the binding of ACE2, CD147, and NRP1 entry factors. Furthermore, exposure to the Spike protein alone proved sufficient to disrupt their homeostasis and induce neurotoxic effects, potentially contributing to the onset of long-COVID symptoms.
Collapse
Affiliation(s)
- Gioia Cappelletti
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Lorenzo Brambilla
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Sergio Strizzi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Fiona Limanaqi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Valentina Melzi
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Mafalda Rizzuti
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Monica Nizzardo
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Irma Saulle
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Daria Trabattoni
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Stefania Corti
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience SectionUniversity of MilanMilanItaly
- Neuromuscular and Rare Diseases Unit, Department of NeuroscienceFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Mario Clerici
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Don C. Gnocchi FoundationIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) FoundationMilanItaly
| | - Mara Biasin
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| |
Collapse
|
2
|
Ferreira RR, Martins RB, Pires I, Marques BL, Costa KC, Lirio PH, Scomparin DS, Scarante FF, Batah SS, Hallak JE, Crippa JA, Rodrigues LC, Tostes RC, Fabro AT, Arruda E, Campos AC. Cardiovascular and kidney diseases are positively associated with neuroinflammation and reduced brain-derived neurotrophic factor in patients with severe COVID-19. Brain Behav Immun Health 2024; 41:100855. [PMID: 39391797 PMCID: PMC11466569 DOI: 10.1016/j.bbih.2024.100855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/25/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Even though respiratory dysfunctions are the primary symptom associated with SARS-CoV-2 infection, cerebrovascular events, and neurological symptoms are described in many patients. However, the connection between the neuroimmune profile and the lung's inflammatory condition during COVID-19 and its association with the neurological symptoms reported by COVID-19 patients still needs further exploration. The present study characterizes the SARS-CoV-2 infectivity profile in postmortem nervous and lung tissue samples of patients who died due to severe COVID-19, and the pro-inflammatory factors present in both nervous and lung tissue samples, via a proteomic profiling array. Additionally, Brain-Derived Neurotrophic Factor (BDNF) levels and intracellular pathways related to neuroplasticity/neuroprotection were assessed in the samples. Out of the 16 samples analyzed, all samples but 1 were positive for the viral genome (genes E or N2, but only 3.9% presented E and N2) in the olfactory brain pathway. The E or N2 gene were also detected in all lung samples, with 43.7% of the samples being positive for the E and N2 genes. In the E/N2 positive brain samples, the Spike protein of SARS-CoV-2 co-localized with TUJ-1+ (neuron-specific class III beta-tubulin) and GFAP+ (glial fibrillary acidic protein) astrocytes. IL-6, but not IL-10, expression was markedly higher in most nervous tissue samples compared to the lung specimens. While intracellular adhesion molecule-1 (ICAM-1), interleukin-8 (IL-8), macrophage migration inhibitory factor (MIF), and plasminogen activator inhibitor 1 (PAI-1) were increased in lung samples from SARS-Cov-2 patients, only MIF and IL-18 were detected in nervous tissue samples. Correlation analysis suggested that high levels of IL-6 are followed by increased levels of IL-10 in the brain, but not in lung samples. Our analysis also demonstrated that the presence of comorbidities, such as cardiovascular disease, hypertension, and hypothyroidism, is associated with neuroinflammation, while chronic kidney conditions predict the presence of neurological symptoms, which correlate with lower levels of BDNF in the brain samples. Our results corroborate the hypothesis that a pro-inflammatory state might further impair neural homeostasis and induce brain abnormalities found in COVID-19 patients.
Collapse
Affiliation(s)
- Rafael R. Ferreira
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Ronaldo B. Martins
- Department of Cell and Molecular Biology, Ribeirão Preto School of Medicine, University of Sao Paulo, Ribeirão Preto, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, São Paulo, Brazil
| | - Isabela Pires
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Bruno L. Marques
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Karla C.M. Costa
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Pedro H.C. Lirio
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Davi S. Scomparin
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Franciele F. Scarante
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Sabrina S. Batah
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, 3900, Bandeirantes Avenue - Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, Brazil
| | - Jaime E.C. Hallak
- Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirão Preto, University of São Paulo, 2650, Tenente Catão Roxo Street - Monte Alegre, Ribeirão Preto – SP- Brazil, 14051-140, São Paulo, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT TM) - CNPQ/FAPESP/CAPES - Brazil
| | - Jose A. Crippa
- Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirão Preto, University of São Paulo, 2650, Tenente Catão Roxo Street - Monte Alegre, Ribeirão Preto – SP- Brazil, 14051-140, São Paulo, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT TM) - CNPQ/FAPESP/CAPES - Brazil
| | - Livia C.M. Rodrigues
- Department of Physiological Sciences, Health Sciences Center, Universidade Federal do Espírito Santo, Vitória 1468, Marechal Campos Avenue - Maruípe, Vitória, ES, 29047-105, Brazil
| | - Rita C. Tostes
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Alexandre T. Fabro
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, 3900, Bandeirantes Avenue - Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, Brazil
| | - Eurico Arruda
- Department of Cell and Molecular Biology, Ribeirão Preto School of Medicine, University of Sao Paulo, Ribeirão Preto, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, São Paulo, Brazil
| | - Alline C. Campos
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
- National Institute of Science and Technology for Translational Medicine (INCT TM) - CNPQ/FAPESP/CAPES - Brazil
| |
Collapse
|
3
|
Eisenreich W, Leberfing J, Rudel T, Heesemann J, Goebel W. Interactions of SARS-CoV-2 with Human Target Cells-A Metabolic View. Int J Mol Sci 2024; 25:9977. [PMID: 39337465 PMCID: PMC11432161 DOI: 10.3390/ijms25189977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Viruses are obligate intracellular parasites, and they exploit the cellular pathways and resources of their respective host cells to survive and successfully multiply. The strategies of viruses concerning how to take advantage of the metabolic capabilities of host cells for their own replication can vary considerably. The most common metabolic alterations triggered by viruses affect the central carbon metabolism of infected host cells, in particular glycolysis, the pentose phosphate pathway, and the tricarboxylic acid cycle. The upregulation of these processes is aimed to increase the supply of nucleotides, amino acids, and lipids since these metabolic products are crucial for efficient viral proliferation. In detail, however, this manipulation may affect multiple sites and regulatory mechanisms of host-cell metabolism, depending not only on the specific viruses but also on the type of infected host cells. In this review, we report metabolic situations and reprogramming in different human host cells, tissues, and organs that are favorable for acute and persistent SARS-CoV-2 infection. This knowledge may be fundamental for the development of host-directed therapies.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Structural Membrane Biochemistry, Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85747 Garching, Germany;
| | - Julian Leberfing
- Structural Membrane Biochemistry, Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85747 Garching, Germany;
| | - Thomas Rudel
- Chair of Microbiology, Biocenter, University of Würzburg, 97074 Würzburg, Germany;
| | - Jürgen Heesemann
- Max von Pettenkofer Institute, Ludwig Maximilian University of Munich, 80336 München, Germany; (J.H.); (W.G.)
| | - Werner Goebel
- Max von Pettenkofer Institute, Ludwig Maximilian University of Munich, 80336 München, Germany; (J.H.); (W.G.)
| |
Collapse
|
4
|
Pavlou A, Mulenge F, Gern OL, Busker LM, Greimel E, Waltl I, Kalinke U. Orchestration of antiviral responses within the infected central nervous system. Cell Mol Immunol 2024; 21:943-958. [PMID: 38997413 PMCID: PMC11364666 DOI: 10.1038/s41423-024-01181-7] [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: 03/29/2024] [Accepted: 05/05/2024] [Indexed: 07/14/2024] Open
Abstract
Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.
Collapse
Affiliation(s)
- Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Elisabeth Greimel
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
5
|
Kim HS, Xiao Y, Chen X, He S, Im J, Willner MJ, Finlayson MO, Xu C, Zhu H, Choi SJ, Mosharov EV, Kim H, Xu B, Leong KW. Chronic Opioid Treatment Arrests Neurodevelopment and Alters Synaptic Activity in Human Midbrain Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400847. [PMID: 38549185 PMCID: PMC11151039 DOI: 10.1002/advs.202400847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Indexed: 06/06/2024]
Abstract
Understanding the impact of long-term opioid exposure on the embryonic brain is critical due to the surging number of pregnant mothers with opioid dependency. However, this has been limited by human brain inaccessibility and cross-species differences in animal models. Here, a human midbrain model is established that uses hiPSC-derived midbrain organoids to assess cell-type-specific responses to acute and chronic fentanyl treatment and fentanyl withdrawal. Single-cell mRNA sequencing of 25,510 cells from organoids in different treatment groups reveals that chronic fentanyl treatment arrests neuronal subtype specification during early midbrain development and alters synaptic activity and neuron projection. In contrast, acute fentanyl treatment increases dopamine release but does not significantly alter gene expression related to cell lineage development. These results provide the first examination of the effects of opioid exposure on human midbrain development at the single-cell level.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Yang Xiao
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Xuejing Chen
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Department of PhysicsTsinghua UniversityBeijing100084China
| | - Siyu He
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Jongwon Im
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Moshe J. Willner
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Michael O. Finlayson
- Single Cell Analysis CoreJP Sulzberger Columbia Genome CenterColumbia University Irving Medical CenterNew YorkNY10032USA
| | - Cong Xu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Huixiang Zhu
- Department of PsychiatryColumbia University Medical CenterNew YorkNY10032USA
| | - Se Joon Choi
- Department of PsychiatryColumbia University Medical CenterNew YorkNY10032USA
- Division of Molecular TherapeuticsNew York State Psychiatric InstituteNew YorkNY10032USA
| | - Eugene V. Mosharov
- Department of PsychiatryColumbia University Medical CenterNew YorkNY10032USA
- Division of Molecular TherapeuticsNew York State Psychiatric InstituteNew YorkNY10032USA
| | - Hae‐Won Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Bin Xu
- Department of PsychiatryColumbia University Medical CenterNew YorkNY10032USA
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Department of Systems BiologyColumbia University Irving Medical CenterNew YorkNY10032USA
| |
Collapse
|
6
|
Shen Q, Zhou YH, Zhou YQ. A prospects tool in virus research: Analyzing the applications of organoids in virus studies. Acta Trop 2024; 254:107182. [PMID: 38479469 DOI: 10.1016/j.actatropica.2024.107182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/23/2024] [Accepted: 03/10/2024] [Indexed: 04/28/2024]
Abstract
Organoids have emerged as a powerful tool for understanding the biology of the respiratory, digestive, nervous as well as urinary system, investigating infections, and developing new therapies. This article reviews recent progress in the development of organoid and advancements in virus research. The potential applications of these models in studying virul infections, pathogenesis, and antiviral drug discovery are discussed.
Collapse
Affiliation(s)
- Qi Shen
- Institute of Microbiology Laboratory, Shanghai Municipal Center for Disease Control and Prevention, Shanghai 20036, China; Institute of Microbiology Laboratory, Shanghai Institute of Preventive Medicine, Shanghai 20036, China
| | - Yu-Han Zhou
- College of Public Health, Jilin University, Changchun 130021, China
| | - Yan-Qiu Zhou
- Institute of Microbiology Laboratory, Shanghai Municipal Center for Disease Control and Prevention, Shanghai 20036, China; Institute of Microbiology Laboratory, Shanghai Institute of Preventive Medicine, Shanghai 20036, China.
| |
Collapse
|
7
|
Ariza D, Castellar-Visbal L, Marquina M, Rivera-Porras D, Galbán N, Santeliz R, Gutiérrez-Rey M, Parra H, Vargas-Manotas J, Torres W, Quintana-Espinosa L, Manzano A, Cudris-Torres L, Bermúdez V. COVID-19: Unveiling the Neuropsychiatric Maze-From Acute to Long-Term Manifestations. Biomedicines 2024; 12:1147. [PMID: 38927354 PMCID: PMC11200893 DOI: 10.3390/biomedicines12061147] [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: 04/17/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
The SARS-CoV-2 virus has spread rapidly despite implementing strategies to reduce its transmission. The disease caused by this virus has been associated with a diverse range of symptoms, including common neurological manifestations such as dysgeusia, anosmia, and myalgias. Additionally, numerous cases of severe neurological complications associated with this disease have been reported, including encephalitis, stroke, seizures, and Guillain-Barré syndrome, among others. Given the high prevalence of neurological manifestations in this disease, the objective of this review is to analyze the mechanisms by which this virus can affect the nervous system, from its direct invasion to aberrant activation of the immune system and other mechanisms involved in the symptoms, including neuropsychiatric manifestations, to gain a better understanding of the disease and thus facilitate the search for effective therapeutic strategies.
Collapse
Affiliation(s)
- Daniela Ariza
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (D.A.); (M.M.); (N.G.); (R.S.); (H.P.); (W.T.); (A.M.)
| | - Lily Castellar-Visbal
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla 080001, Colombia; (L.C.-V.); (M.G.-R.); (J.V.-M.); (L.Q.-E.)
| | - Maria Marquina
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (D.A.); (M.M.); (N.G.); (R.S.); (H.P.); (W.T.); (A.M.)
| | - Diego Rivera-Porras
- Universidad Simón Bolívar, Facultad de Ciencias Jurídicas y Sociales, Centro de Investigación en Estudios Fronterizos, Cúcuta 540001, Colombia;
| | - Nestor Galbán
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (D.A.); (M.M.); (N.G.); (R.S.); (H.P.); (W.T.); (A.M.)
| | - Raquel Santeliz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (D.A.); (M.M.); (N.G.); (R.S.); (H.P.); (W.T.); (A.M.)
| | - Melissa Gutiérrez-Rey
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla 080001, Colombia; (L.C.-V.); (M.G.-R.); (J.V.-M.); (L.Q.-E.)
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (D.A.); (M.M.); (N.G.); (R.S.); (H.P.); (W.T.); (A.M.)
| | - José Vargas-Manotas
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla 080001, Colombia; (L.C.-V.); (M.G.-R.); (J.V.-M.); (L.Q.-E.)
| | - Wheeler Torres
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (D.A.); (M.M.); (N.G.); (R.S.); (H.P.); (W.T.); (A.M.)
| | - Laura Quintana-Espinosa
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla 080001, Colombia; (L.C.-V.); (M.G.-R.); (J.V.-M.); (L.Q.-E.)
| | - Alexander Manzano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, Universidad del Zulia, Maracaibo 4002, Venezuela; (D.A.); (M.M.); (N.G.); (R.S.); (H.P.); (W.T.); (A.M.)
| | - Lorena Cudris-Torres
- Departamento de Ciencias Sociales, Universidad de la Costa, Barranquilla 080001, Colombia;
| | - Valmore Bermúdez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla 080001, Colombia; (L.C.-V.); (M.G.-R.); (J.V.-M.); (L.Q.-E.)
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Centro de Investigaciones en Ciencias de la Vida, Barranquilla 080001, Colombia
| |
Collapse
|
8
|
Chen W, Jiang X, Liang W, Bai H, Xu M, Liu Z, Yi L, Liu Y, Huang Y, Zhang Y, Xu L, Xie B, Zhang N, Yu J, Lu J, Xiao H, Li X. SARS-CoV-2 Omicron Variants Show Attenuated Neurovirulence Compared with the Wild-Type Strain in Elderly Human Brain Spheroids. RESEARCH (WASHINGTON, D.C.) 2024; 7:0376. [PMID: 38741604 PMCID: PMC11089278 DOI: 10.34133/research.0376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/13/2024] [Indexed: 05/16/2024]
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 Omicron variants still causes neurological complications in elderly individuals. However, whether and how aging brains are affected by Omicron variants in terms of neuroinvasiveness and neurovirulence are unknown. Here, we utilize resected paracarcinoma brain tissue from elderly individuals to generate primary brain spheroids (BSs) for investigating the replication capability of live wild-type (WT) strain and Omicron (BA.1/BA.2), as well as the mechanisms underlying their neurobiological effects. We find that both WT and Omicron BA.1/BA.2 are able to enter BSs but weakly replicate. There is no difference between Omicron BA.1/BA.2 and WT strains in neurotropism in aging BSs. However, Omicron BA.1/BA.2 exhibits ameliorating neurological damage. Transcriptional profiling indicates that Omicron BA.1/BA.2 induces a lower neuroinflammatory response than WT strain in elderly BSs, suggesting a mechanistic explanation for their attenuated neuropathogenicity. Moreover, we find that both Omicron BA.1/BA.2 and WT strain infections disrupt neural network activity associated with neurodegenerative disorders by causing neuron degeneration and amyloid-β deposition in elderly BSs. These results uncover Omicron-specific mechanisms and cellular immune responses associated with severe acute respiratory syndrome coronavirus 2-induced neurological complications.
Collapse
Affiliation(s)
- Weikang Chen
- Department of Neurosurgery,
The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Xiaobing Jiang
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China,
Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou 510000, China
| | - Wei Liang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Haojie Bai
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Mingze Xu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Zhe Liu
- Guangdong Provincial Institution of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510000, China
| | - Lina Yi
- Guangdong Provincial Institution of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510000, China
| | - Yanming Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Yanxia Huang
- Department of Neurosurgery,
The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Yongxin Zhang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Lixia Xu
- Department of Oncology,
The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Baoshu Xie
- Department of Neurosurgery,
The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Nu Zhang
- Department of Neurosurgery,
The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Jun Yu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
- Department of Medicine and Therapeutics and Institute of Digestive Disease, State Key Laboratory of Digestive Disease,
The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Jing Lu
- Guangdong Provincial Institution of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 510000, China
| | - Haipeng Xiao
- Department of Endocrinology,
The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Xiaoxing Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, China
| |
Collapse
|
9
|
Vavougios GD, Mavridis T, Doskas T, Papaggeli O, Foka P, Hadjigeorgiou G. SARS-CoV-2-Induced Type I Interferon Signaling Dysregulation in Olfactory Networks Implications for Alzheimer's Disease. Curr Issues Mol Biol 2024; 46:4565-4579. [PMID: 38785545 PMCID: PMC11119810 DOI: 10.3390/cimb46050277] [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: 01/10/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Type I interferon signaling (IFN-I) perturbations are major drivers of COVID-19. Dysregulated IFN-I in the brain, however, has been linked to both reduced cognitive resilience and neurodegenerative diseases such as Alzheimer's. Previous works from our group have proposed a model where peripheral induction of IFN-I may be relayed to the CNS, even in the absence of fulminant infection. The aim of our study was to identify significantly enriched IFN-I signatures and genes along the transolfactory route, utilizing published datasets of the nasal mucosa and olfactory bulb amygdala transcriptomes of COVID-19 patients. We furthermore sought to identify these IFN-I signature gene networks associated with Alzheimer's disease pathology and risk. Gene expression data involving the nasal epithelium, olfactory bulb, and amygdala of COVID-19 patients and transcriptomic data from Alzheimer's disease patients were scrutinized for enriched Type I interferon pathways. Gene set enrichment analyses and gene-Venn approaches were used to determine genes in IFN-I enriched signatures. The Agora web resource was used to identify genes in IFN-I signatures associated with Alzheimer's disease risk based on its aggregated multi-omic data. For all analyses, false discovery rates (FDR) <0.05 were considered statistically significant. Pathways associated with type I interferon signaling were found in all samples tested. Each type I interferon signature was enriched by IFITM and OAS family genes. A 14-gene signature was associated with COVID-19 CNS and the response to Alzheimer's disease pathology, whereas nine genes were associated with increased risk for Alzheimer's disease based on Agora. Our study provides further support to a type I interferon signaling dysregulation along the extended olfactory network as reconstructed herein, ranging from the nasal epithelium and extending to the amygdala. We furthermore identify the 14 genes implicated in this dysregulated pathway with Alzheimer's disease pathology, among which HLA-C, HLA-B, HLA-A, PSMB8, IFITM3, HLA-E, IFITM1, OAS2, and MX1 as genes with associated conferring increased risk for the latter. Further research into its druggability by IFNb therapeutics may be warranted.
Collapse
Affiliation(s)
- George D. Vavougios
- Department of Neurology, Medical School, University of Cyprus, Nicosia 1678, Cyprus
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital, Dublin, Incorporating the National Children’s Hospital (AMNCH), D24 NR0A Dublin, Ireland;
| | | | - Olga Papaggeli
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 115 21 Athens, Greece; (O.P.); (P.F.)
| | - Pelagia Foka
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 115 21 Athens, Greece; (O.P.); (P.F.)
| | | |
Collapse
|
10
|
Vavougios GD, Tseriotis VS, Liampas A, Mavridis T, de Erausquin GA, Hadjigeorgiou G. Type I interferon signaling, cognition and neurodegeneration following COVID-19: update on a mechanistic pathogenetic model with implications for Alzheimer's disease. Front Hum Neurosci 2024; 18:1352118. [PMID: 38562226 PMCID: PMC10982434 DOI: 10.3389/fnhum.2024.1352118] [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: 12/07/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
COVID-19's effects on the human brain reveal a multifactorial impact on cognition and the potential to inflict lasting neuronal damage. Type I interferon signaling, a pathway that represents our defense against pathogens, is primarily affected by COVID-19. Type I interferon signaling, however, is known to mediate cognitive dysfunction upon its dysregulation following synaptopathy, microgliosis and neuronal damage. In previous studies, we proposed a model of outside-in dysregulation of tonic IFN-I signaling in the brain following a COVID-19. This disruption would be mediated by the crosstalk between central and peripheral immunity, and could potentially establish feed-forward IFN-I dysregulation leading to neuroinflammation and potentially, neurodegeneration. We proposed that for the CNS, the second-order mediators would be intrinsic disease-associated molecular patterns (DAMPs) such as proteopathic seeds, without the requirement of neuroinvasion to sustain inflammation. Selective vulnerability of neurogenesis sites to IFN-I dysregulation would then lead to clinical manifestations such as anosmia and cognitive impairment. Since the inception of our model at the beginning of the pandemic, a growing body of studies has provided further evidence for the effects of SARS-CoV-2 infection on the human CNS and cognition. Several preclinical and clinical studies have displayed IFN-I dysregulation and tauopathy in gene expression and neuropathological data in new cases, correspondingly. Furthermore, neurodegeneration identified with a predilection for the extended olfactory network furthermore supports the neuroanatomical concept of our model, and its independence from fulminant neuroinvasion and encephalitis as a cause of CNS damage. In this perspective, we summarize the data on IFN-I as a plausible mechanism of cognitive impairment in this setting, and its potential contribution to Alzheimer's disease and its interplay with COVID-19.
Collapse
Affiliation(s)
- George D. Vavougios
- Department of Neurology, Medical School, University of Cyprus, Lefkosia, Cyprus
| | | | - Andreas Liampas
- Department of Neurology, Medical School, University of Cyprus, Lefkosia, Cyprus
| | - Theodore Mavridis
- Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital Dublin, Incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | - Gabriel A. de Erausquin
- Laboratory of Brain Development, Modulation and Repair, The Glenn Biggs Institute of Alzheimer's and Neurodegenerative Disorders, Joe R. and Teresa Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | | |
Collapse
|
11
|
Wang M, Wang J, Ren Y, Lu L, Xiong W, Li L, Xu S, Tang M, Yuan Y, Xie Y, Li W, Chen L, Zhou D, Ying B, Li J. Current clinical findings of acute neurological syndromes after SARS-CoV-2 infection. MedComm (Beijing) 2024; 5:e508. [PMID: 38463395 PMCID: PMC10924641 DOI: 10.1002/mco2.508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 03/12/2024] Open
Abstract
Neuro-COVID, a condition marked by persistent symptoms post-COVID-19 infection, notably affects various organs, with a particular focus on the central nervous system (CNS). Despite scant evidence of SARS-CoV-2 invasion in the CNS, the increasing incidence of Neuro-COVID cases indicates the onset of acute neurological symptoms early in infection. The Omicron variant, distinguished by heightened neurotropism, penetrates the CNS via the olfactory bulb. This direct invasion induces inflammation and neuronal damage, emphasizing the need for vigilance regarding potential neurological complications. Our multicenter study represents a groundbreaking revelation, documenting the definite presence of SARS-CoV-2 in the cerebrospinal fluid (CSF) of a significant proportion of Neuro-COVID patients. Furthermore, notable differences emerged between RNA-CSF-positive and negative patients, encompassing aspects such as blood-brain barrier integrity, extent of neuronal damage, and the activation status of inflammation. Despite inherent limitations, this research provides pivotal insights into the intricate interplay between SARS-CoV-2 and the CNS, underscoring the necessity for ongoing research to fully comprehend the virus's enduring effects on the CNS. The findings underscore the urgency of continuous investigation Neuro-COVID to unravel the complexities of this relationship, and pivotal in addressing the long-term consequences of COVID-19 on neurological health.
Collapse
Affiliation(s)
- Minjin Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Department of Laboratory MedicineWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐inspired TechnologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Jierui Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐inspired TechnologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Yan Ren
- Department of Laboratory MedicineWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Lu Lu
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐inspired TechnologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Weixi Xiong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐inspired TechnologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Lifeng Li
- Genskey Medical biotechnology Company LimitedBeijingChina
| | - Songtao Xu
- National Institute for Viral Disease Control and PreventionChinese Center for Disease Control and PreventionBeijingChina
| | - Meng Tang
- Department of Laboratory MedicineWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Yushang Yuan
- Department of Laboratory MedicineWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Yi Xie
- Department of Laboratory MedicineWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Weimin Li
- Department of Respiratory and Critical Care MedicineWest China HospitalSichuan UniversityChengduSichuanChina
| | - Lei Chen
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐inspired TechnologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐inspired TechnologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Binwu Ying
- Department of Laboratory MedicineWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Jinmei Li
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐inspired TechnologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| |
Collapse
|
12
|
Dos Reis VP, Cirksena K, Rybak-Wolf A, Seeger B, Herker E, Gerold G. 3D Spheroid and Organoid Models to Study Neuroinfection of RNA Viruses. Methods Mol Biol 2024; 2824:409-424. [PMID: 39039427 DOI: 10.1007/978-1-0716-3926-9_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Three-dimensional culture models of the brain enable the study of neuroinfection in the context of a complex interconnected cell matrix. Depending on the differentiation status of the neural cells, two models exist: 3D spheroids also called neurospheres and cerebral organoids. Here, we describe the preparation of 3D spheroids and cerebral organoids and give an outlook on their usage to study Rift Valley fever virus and other neurotropic viruses.
Collapse
Affiliation(s)
| | - Karsten Cirksena
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Agnieszka Rybak-Wolf
- Organoid Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.
| | - Bettina Seeger
- Institute for Food Quality and Safety, University of Veterinary Medicine Hannover, Hannover, Germany.
| | - Eva Herker
- Institute of Virology, Philipps-University Marburg, Marburg, Germany.
| | - Gisa Gerold
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany.
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden.
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden.
| |
Collapse
|
13
|
Li M, Yuan Y, Zou T, Hou Z, Jin L, Wang B. Development trends of human organoid-based COVID-19 research based on bibliometric analysis. Cell Prolif 2023; 56:e13496. [PMID: 37218396 PMCID: PMC10693193 DOI: 10.1111/cpr.13496] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/13/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), a global pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has posed a catastrophic threat to human health worldwide. Human stem cell-derived organoids serve as a promising platform for exploring SARS-CoV-2 infection. Several review articles have summarized the application of human organoids in COVID-19, but the research status and development trend of this field have seldom been systematically and comprehensively studied. In this review, we use bibliometric analysis method to identify the characteristics of organoid-based COVID-19 research. First, an annual trend of publications and citations, the most contributing countries or regions and organizations, co-citation analysis of references and sources and research hotspots are determined. Next, systematical summaries of organoid applications in investigating the pathology of SARS-CoV-2 infection, vaccine development and drug discovery, are provided. Lastly, the current challenges and future considerations of this field are discussed. The present study will provide an objective angle to identify the current trend and give novel insights for directing the future development of human organoid applications in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
- Southwest Hospital/Southwest Eye HospitalThird Military Medical University (Army Medical University)ChongqingChina
| | - Yuhan Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Ting Zou
- Southwest Hospital/Southwest Eye HospitalThird Military Medical University (Army Medical University)ChongqingChina
| | - Zongkun Hou
- School of Basic Medical Sciences/School of Biology and Engineering (School of Modern Industry for Health and Medicine)Guizhou Medical UniversityGuiyangChina
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| |
Collapse
|
14
|
Mignolet M, Gilloteaux J, Halloin N, Gueibe M, Willemart K, De Swert K, Bielarz V, Suain V, Pastushenko I, Gillet NA, Nicaise C. Viral Entry Inhibitors Protect against SARS-CoV-2-Induced Neurite Shortening in Differentiated SH-SY5Y Cells. Viruses 2023; 15:2020. [PMID: 37896797 PMCID: PMC10611151 DOI: 10.3390/v15102020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
The utility of human neuroblastoma cell lines as in vitro model to study neuro-invasiveness and neuro-virulence of SARS-CoV-2 has been demonstrated by our laboratory and others. The aim of this report is to further characterize the associated cellular responses caused by a pre-alpha SARS-CoV-2 strain on differentiated SH-SY5Y and to prevent its cytopathic effect by using a set of entry inhibitors. The susceptibility of SH-SY5Y to SARS-CoV-2 was confirmed at high multiplicity-of-infection, without viral replication or release. Infection caused a reduction in the length of neuritic processes, occurrence of plasma membrane blebs, cell clustering, and changes in lipid droplets electron density. No changes in the expression of cytoskeletal proteins, such as tubulins or tau, could explain neurite shortening. To counteract the toxic effect on neurites, entry inhibitors targeting TMPRSS2, ACE2, NRP1 receptors, and Spike RBD were co-incubated with the viral inoculum. The neurite shortening could be prevented by the highest concentration of camostat mesylate, anti-RBD antibody, and NRP1 inhibitor, but not by soluble ACE2. According to the degree of entry inhibition, the average amount of intracellular viral RNA was negatively correlated to neurite length. This study demonstrated that targeting specific SARS-CoV-2 host receptors could reverse its neurocytopathic effect on SH-SY5Y.
Collapse
Affiliation(s)
- Margaux Mignolet
- URPhyM, NARILIS, Faculté de Médecine, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.M.); (J.G.); (N.H.); (M.G.); (K.D.S.); (V.B.)
| | - Jacques Gilloteaux
- URPhyM, NARILIS, Faculté de Médecine, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.M.); (J.G.); (N.H.); (M.G.); (K.D.S.); (V.B.)
- Department of Anatomical Sciences, St George’s University School of Medicine, Newcastle upon Tyne NE1 JG8, UK
| | - Nicolas Halloin
- URPhyM, NARILIS, Faculté de Médecine, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.M.); (J.G.); (N.H.); (M.G.); (K.D.S.); (V.B.)
| | - Matthieu Gueibe
- URPhyM, NARILIS, Faculté de Médecine, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.M.); (J.G.); (N.H.); (M.G.); (K.D.S.); (V.B.)
| | - Kévin Willemart
- URVI, NARILIS, Faculté des Sciences, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (K.W.); (N.A.G.)
| | - Kathleen De Swert
- URPhyM, NARILIS, Faculté de Médecine, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.M.); (J.G.); (N.H.); (M.G.); (K.D.S.); (V.B.)
| | - Valéry Bielarz
- URPhyM, NARILIS, Faculté de Médecine, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.M.); (J.G.); (N.H.); (M.G.); (K.D.S.); (V.B.)
| | - Valérie Suain
- Laboratoire d’Histologie Générale, Faculté de Médecine, Université Libre de Bruxelles, Route de Lennik 808, 1070 Bruxelles, Belgium; (V.S.); (I.P.)
| | - Ievgenia Pastushenko
- Laboratoire d’Histologie Générale, Faculté de Médecine, Université Libre de Bruxelles, Route de Lennik 808, 1070 Bruxelles, Belgium; (V.S.); (I.P.)
| | - Nicolas Albert Gillet
- URVI, NARILIS, Faculté des Sciences, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (K.W.); (N.A.G.)
| | - Charles Nicaise
- URPhyM, NARILIS, Faculté de Médecine, Université de Namur, Rue de Bruxelles 61, 5000 Namur, Belgium; (M.M.); (J.G.); (N.H.); (M.G.); (K.D.S.); (V.B.)
| |
Collapse
|
15
|
Guevara-Ramírez P, Cadena-Ullauri S, Paz-Cruz E, Tamayo-Trujillo R, Ruiz-Pozo VA, Zambrano AK. Role of the gut microbiota in hematologic cancer. Front Microbiol 2023; 14:1185787. [PMID: 37692399 PMCID: PMC10485363 DOI: 10.3389/fmicb.2023.1185787] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Hematologic neoplasms represent 6.5% of all cancers worldwide. They are characterized by the uncontrolled growth of hematopoietic and lymphoid cells and a decreased immune system efficacy. Pathological conditions in hematologic cancer could disrupt the balance of the gut microbiota, potentially promoting the proliferation of opportunistic pathogens. In this review, we highlight studies that analyzed and described the role of gut microbiota in different types of hematologic diseases. For instance, myeloma is often associated with Pseudomonas aeruginosa and Clostridium leptum, while in leukemias, Streptococcus is the most common genus, and Lachnospiraceae and Ruminococcaceae are less prevalent. Lymphoma exhibits a moderate reduction in microbiota diversity. Moreover, certain factors such as delivery mode, diet, and other environmental factors can alter the diversity of the microbiota, leading to dysbiosis. This dysbiosis may inhibit the immune response and increase susceptibility to cancer. A comprehensive analysis of microbiota-cancer interactions may be useful for disease management and provide valuable information on host-microbiota dynamics, as well as the possible use of microbiota as a distinguishable marker for cancer progression.
Collapse
|
16
|
Tso WWY, Kwan MYW, Kwok JSY, Tsang JOL, Yip CCY, Leung LK, Li C, Wang Y, Chow MSC, Tsang AMC, Chim S, Chow CY, Ho ACC, Chan SHS, Tai SM, Lee WC, Chan VCM, Yau EKC, Sun JKL, Chow HM, Lau YL, Ip P, Chan JFW. Clinical characteristics of unvaccinated or incompletely vaccinated children with neurological manifestations due to SARS-CoV-2 Omicron infection. J Med Virol 2023; 95:e28895. [PMID: 37403902 DOI: 10.1002/jmv.28895] [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: 04/25/2023] [Revised: 06/06/2023] [Accepted: 06/11/2023] [Indexed: 07/06/2023]
Abstract
Omicron generally causes milder disease than previous strains of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), especially in fully vaccinated individuals. However, incompletely vaccinated children may develop Omicron-related complications such as those affecting the central nervous system. To characterize the spectrum of clinical manifestations of neuro-COVID and to identify potential biomarkers associated with clinical outcomes, we recruited 15 children hospitalized for Omicron-related neurological manifestations in three hospitals in Hong Kong (9 boys and 6 girls aged 1-13 years). All were unvaccinated or incompletely vaccinated. Fourteen (93.3%) were admitted for convulsion, including benign febrile seizure (n = 7), complex febrile seizure (n = 2), seizure with fever (n = 3), and recurrent breakthrough seizure (n = 2), and the remaining nonconvulsive patient developed encephalopathic state with impaired consciousness. None of the seven children with benign febrile seizure and six of eight children with other neurological manifestations had residual deficits at 9-month follow-up. SARS-CoV-2 RNA was undetectable in the cerebrospinal fluid (CSF) specimens of seven patients who underwent lumbar puncture. Spike-and-wave/sharp waves affecting the frontal lobes were detected in four of seven (57.1%) patients who underwent electroencephalogram. Children with Omicron-related neurological manifestations had significantly higher blood levels of IL-6 (p < 0.001) and CHI3L1 (p = 0.022) than healthy controls, and higher CSF levels of IL-6 (p = 0.002) than children with non-COVID-19-related febrile illnesses. Higher CSF-to-blood ratios of IL-8 and CHI3L1 were associated with longer length of stay, whereas higher ratios of IL-6 and IL-8 were associated with higher blood tau level. The role of CSF:blood ratio of IL-6, IL-8, and CHI3L1 as prognostic markers for neuro-COVID should be further evaluated.
Collapse
Affiliation(s)
- Winnie Wan-Yee Tso
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- State Key Laboratory of Brain & Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Mike Yat-Wah Kwan
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Janette Siu-Yin Kwok
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jessica Oi-Ling Tsang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, China
| | - Cyril Chik-Yan Yip
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Lok-Kan Leung
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Cuixin Li
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yuliang Wang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Mathew Siu-Chun Chow
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anita Man-Ching Tsang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Stella Chim
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Chin-Ying Chow
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Alvin Chi-Chung Ho
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Sophelia Hoi-Shan Chan
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shuk-Mui Tai
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong Special Administrative Region, China
| | - Wing-Cheong Lee
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong Special Administrative Region, China
| | - Victor Chi-Man Chan
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong Special Administrative Region, China
| | - Eric Kin-Cheong Yau
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Jacquelyne Ka-Li Sun
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Hei-Man Chow
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
- Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Yu-Lung Lau
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Patrick Ip
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Sha Tin, Hong Kong Special Administrative Region, China
- Department of Infectious Diseases and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
17
|
Liu Y. Integrative network pharmacology and in silico analyses identify the anti-omicron SARS-CoV-2 potential of eugenol. Heliyon 2023; 9:e13853. [PMID: 36845041 PMCID: PMC9937729 DOI: 10.1016/j.heliyon.2023.e13853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Eugenol as a natural product is the source of isoniazid, and purified eugenol is extensively used in the cosmetics industry and the productive processes of edible spices. Accumulating evidence suggested that eugenol exerted potent anti-microorganism and anti-inflammation effects. Application of eugenol effectively reduced the risk of atherosclerosis, arterial embolism, and Type 2 diabetes. A previous study confirmed that treatment with eugenol attenuated lung inflammation and improved heart functions in SARS-CoV-2 spike S1-intoxicated mice. In addition to the study, based on a series of public datasets, computational analyses were conducted to characterize the acting targets of eugenol and the functional roles of these targets in COVID-19. The binding capacities of eugenol to conservative sites of SARS-CoV-2 like RNA-dependent RNA polymerase (RdRp) and mutable site as spike (S) protein, were calculated by using molecular docking following the molecular dynamics simulation with RMSD, RMSF, and MM-GBSA methods. The results of network pharmacology indicated that six targets, including PLAT, HMOX1, NUP88, CTSL, ITGB1 andTMPRSS2 were eugenol-SARS-CoV-2 interacting proteins. The omics results of in-silico study further implicated that eugenol increased the expression of SCARB1, HMOX1 and GDF15, especially HMOX1, which were confirmed the potential interacting targets between eugenol and SARS-CoV-2 antigens. Enrichment analyses indicated that eugenol exerted extensive biological effects such as regulating immune infiltration of macrophage, lipid localization, monooxyenase activity, iron ion binding and PPAR signaling. The results of the integrated analysis of eugenol targets and immunotranscription profile of COVID-19 cases shows that eugenol also plays an important role in strengthen of immunologic functions and regulating cytokine signaling. As a complement to the integrated analysis, the results of molecular docking indicated the potential binding interactions between eugenol and four proteins relating to cytokine production/release and the function of T type lymphocytes, including human TLR-4, TCR, NF-κB, JNK and AP-1. Furthermore, results of molecular docking and molecular dynamics (100ns) simulations implicated that stimulated modification of eugenol to the SARS-CoV-2 Omicron Spike-ACE2 complex, especially for human ACE2, and the molecular interaction of eugenol to SARS-CoV-2 RdRp, were no less favorable than two positive controls, molnupiravir and nilotinib. Dynamics (200ns) simulations indicated that the binding capacities and stabilities of eugenol to finger subdomain of RdRp is no less than molnupiravir. However, the simulated binding capacity of eugenol to SARS-CoV-2 wild type RBD and Omicron mutant RBD were less than nilotinib. Eugenol was predicted to have more favor LD50 value and lower cytotoxicity than two positive controls, and eugenol can pass through the blood-brain barrier (BBB). In a brief, eugenol is helpful for attenuating systemic inflammation induced by SARS-CoV-2 infection, due to the direct interaction of eugenol to SARS-CoV-2 proteins and extensive bio-manipulation of pro-inflammatory factors. This study carefully suggests eugenol is a candidate compound of developing drugs and supplement agents against SARS-CoV-2 and its Omicron variants.
Collapse
Affiliation(s)
- Yang Liu
- Graduated Student of Harbin Medical University, Cardiology. Baojian Road105, Nangang Distinct, Harbin, Heilongjiang, China
| |
Collapse
|