1
|
Wang X, Kong Y. Advancing mechanistic understanding of post-acute sequelae of Ebola virus disease. THE LANCET. MICROBE 2025:101139. [PMID: 40258383 DOI: 10.1016/j.lanmic.2025.101139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/23/2025]
Affiliation(s)
- Xinyue Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yaxian Kong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Subbarayan K, Bieber H, Massa C, Rodríguez FAE, Hossain SMAA, Neuder L, Wahbi W, Salo T, Tretbar S, Al-Samadi A, Seliger B. Link of TMPRSS2 expression with tumor immunogenicity and response to immune checkpoint inhibitors in cancers. J Transl Med 2025; 23:294. [PMID: 40055791 PMCID: PMC11887338 DOI: 10.1186/s12967-025-06177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND SARS-CoV-2 and other viruses rely on the protease function of the TMPRSS2 protein to invade host cells. Despite cancer patients often experience poorer outcomes following SARS-CoV-2 infection, the role of TMPRSS2 in different cancer types has not yet been analyzed in detail. Therefore, the aim of the study was to determine the expression, function and clinical relevance of TMPRSS2 in tumors. METHODS Publicly accessible RNA sequencing data from tumors, adjacent tissues and whole blood samples of COVID-19 patients as well as data from human tumor epithelial and endothelial cells infected with SARS-CoV-2 were analyzed for TMPRSS2 expression and correlated to the expression of immune-relevant genes and clinical parameters. In vitro models of cells transfected with TMPRSS2 (TMPRSS2high), siTMPRSS2 or mock controls (TMPRSS2low cells) were analyzed by qPCR, flow cytometry, ELISA and Western blot for the expression of immune response-relevant molecules. Co-cultures of TMPRSS2 model systems with blood peripheral mononuclear cells were employed to evaluate immune cell migration, cytotoxicity and cytokine release. RESULTS Higher expression levels of TMPRSS2 were found in blood from patients infected with SARS-CoV-2, while TMPRSS2 expression levels significantly varied between the tumor types analyzed. TMPRSS2high tumor cells exhibit increased activity of the interferon (IFN) signal pathway accompanied by an increased expression of class I human leukocyte antigens (HLA-I) and programmed cell death ligand 1 (PD-L1) elevated interleukin 6 (IL-6) secretion and reduced NK cell-mediated cytotoxicity compared to TMPRSS2low mock controls. Treatment with a Janus kinase (JAK) 2 inhibitor or TMPRSS2-specific siRNA decreased TMPRSS2 expression. Co-cultures of the in vitro TMPRSS2 models with peripheral blood mononuclear cells in the presence of the immune checkpoint inhibitor nivolumab resulted in a significantly increased migration and infiltration of immune cells towards TMPRSS2high cells and a reduced release of the innate immunity-related cytokines CCL2 and CCL3. CONCLUSIONS This study provides novel insights into the role of TMPRSS2 in various tumor systems and the impact of SARS-CoV-2 infection on the host immunogenicity via the activation of immune-relevant pathways. These findings were linked to the efficacy of immune checkpoint inhibitor therapy, offering a potential alternative strategy to mitigate the severity of COVID-19.
Collapse
Affiliation(s)
| | - Helena Bieber
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chiara Massa
- Institute of Translational Immunology, Faculty of Health Sciences, Brandenburg Medical School "Theodor Fontane", Brandenburg an der Havel, Germany
| | - Felipe Adonis Escalona Rodríguez
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Center for Protein Studies, Faculty of Biology, University of Havana (UH), Havana, Cuba
- NanoCancer, Molecular Immunology Center (CIM), Havana, Cuba
| | - S M Al Amin Hossain
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Lisa Neuder
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, 90014, Finland
| | - Sandy Tretbar
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Institute of Dentistry, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Institute of Translational Immunology, Faculty of Health Sciences, Brandenburg Medical School "Theodor Fontane", Brandenburg an der Havel, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
| |
Collapse
|
3
|
Codazzi V, Salvatore V, Ragogna F, Marzinotto I, Anselmo A, Baldoni N, Pastore MR, Martinenghi S, Stabilini A, Bosi E, Giustina A, Piemonti L, Libman I, Ismail HM, Redondo MJ, Lampasona V, Monti P, Giovenzana A, Petrelli A. Metabolic, genetic and immunological features of relatives of type 1 diabetes patients with elevated insulin resistance. J Endocrinol Invest 2025; 48:765-775. [PMID: 39656436 PMCID: PMC11876269 DOI: 10.1007/s40618-024-02497-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/01/2024] [Indexed: 03/04/2025]
Abstract
PURPOSE Insulin resistance plays a pivotal role in the preclinical stages of type 1 diabetes (T1D). OBJECTIVE This study aims at exploring the genetic, metabolic, and immunological features associated with insulin resistance among individuals at risk of developing T1D. METHODS We retrospectively selected relatives of individuals with T1D from participants in the TrialNet Pathway to Prevention study. They were categorized into two groups: high-H (n = 27) and low-H (n = 30), based on the upper and lower quartiles of insulin resistance assessed using the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR). Genetic predisposition was determined using the T1D Genetic Risk Score 1 (GRS1). Additionally, glucose control was evaluated through an oral glucose tolerance test and levels of metabolic hormones and inflammatory cytokines were measured in the serum. Flow cytometry analysis was employed to assess frequency and phenotype of islet-specific CD8 T cells. RESULTS While GRS1 were similar between the low-H and high-H groups, high-H individuals displayed a distinct metabolic profile, characterized by compensatory hyperinsulinemia, even while maintaining normoglycemia. Circulating cytokine levels were similar between the two groups. However, immune profiling revealed a central memory and activated profile of GAD65-specific CD8 T cells, along with an increased frequency of insulin-specific CD8 T cells in high-H individuals. The enrichment in insulin-specific CD8 T cells was independent of body mass. CONCLUSION These findings highlight the intricate interplay between insulin resistance, genetic factors, and immune activation in the context of T1D susceptibility, indicating potential connections between insulin resistance and immune responses specific to islet cells.
Collapse
Affiliation(s)
- V Codazzi
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - V Salvatore
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - F Ragogna
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - I Marzinotto
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - A Anselmo
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - N Baldoni
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - M R Pastore
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - S Martinenghi
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - A Stabilini
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - E Bosi
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - A Giustina
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - L Piemonti
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - I Libman
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - H M Ismail
- Indiana University School of Medicine, Indianapolis, USA
| | - M J Redondo
- Texas Children's Hospital, Baylor College of Medicine, Houston, USA
| | - V Lampasona
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - P Monti
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - A Giovenzana
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy
| | - A Petrelli
- IRCCS Ospedale San Raffaele, Via Olgettina 60, Milan, 20132, Italy.
- University of Milan and Pio Albergo Trivulzio, Milan, Italy.
| |
Collapse
|
4
|
Andrade Barboza C, Gonçalves LM, Pereira E, Cruz RD, Andrade Louzada R, Boulina M, Almaça J. SARS-CoV-2 Spike S1 Subunit Triggers Pericyte and Microvascular Dysfunction in Human Pancreatic Islets. Diabetes 2025; 74:355-367. [PMID: 39715591 PMCID: PMC11842606 DOI: 10.2337/db24-0816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
The COVID-19 pandemic has profoundly affected human health; however, the mechanisms underlying its impact on metabolic and vascular systems remain incompletely understood. Clinical evidence suggests that SARS-CoV-2 directly disrupts vascular homeostasis, with perfusion abnormalities observed in various tissues. The pancreatic islet, a key endocrine miniorgan reliant on its microvasculature for optimal function, may be particularly vulnerable. Studies have proposed a link between SARS-CoV-2 infection and islet dysfunction, but the mechanisms remain unclear. Here, we investigated how SARS-CoV-2 spike S1 protein affects human islet microvascular function. Using confocal microscopy and living pancreas slices from organ donors without diabetes, we show that a SARS-CoV-2 spike S1 recombinant protein activates pericytes, key regulators of islet capillary diameter and β-cell function, and induces capillary constriction. These effects are driven by a loss of ACE2 from pericytes' plasma membrane, impairing ACE2 activity and increasing local angiotensin II levels. Our findings highlight islet pericyte dysfunction as a potential contributor to the diabetogenic effects of SARS-CoV-2 and offer new insights into the mechanisms linking COVID-19, vascular dysfunction, and diabetes. ARTICLE HIGHLIGHTS Different components of the renin-angiotensin system are expressed by vascular cells in human pancreatic islets. The islet microvasculature is responsive to vasoactive angiotensin peptides. This pancreatic renin-angiotensin system is targeted upon incubation with a SARS-CoV-2 spike recombinant protein. SARS-CoV-2 spike activates pericytes and constricts capillaries in human islets. Islet vascular dysfunction could contribute to dysglycemia in some patients with COVID-19.
Collapse
Affiliation(s)
- Catarina Andrade Barboza
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Elizabeth Pereira
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
| | - Roxana Diaz Cruz
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Ruy Andrade Louzada
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Maria Boulina
- Diabetes Research Institute, University of Miami Health System, Miami, FL
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Health System, Miami, FL
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
5
|
Lundstrom K. Immunobiology and immunotherapy of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:73-133. [PMID: 40246352 DOI: 10.1016/bs.pmbts.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The SARS-CoV-2 outbreak in late 2019 triggered a major increase in activities related to immunobiology and immunotherapy to cope with and find solutions to end the COVID-19 pandemic. The unprecedented approach to research and development of drugs and vaccines against SARS-CoV-2 has substantially improved the understanding of immunobiology for COVID-19, which can also be applied to other infectious diseases. Major efforts were dedicated to the repurposing of existing antiviral drugs and the development of novel ones. For this reason, numerous approaches to evaluating interferons, immunoglobulins, and cytokine inhibitors have been conducted. Antibody-based therapies, especially employing monoclonal antibodies have also been on the agenda. Cell-based therapies involving dendritic cells, macrophages, and CAR T-cell approaches have been evaluated. Many existing antiviral drugs have been repurposed for COVID-19 and novel formulations have been tested. The extraordinarily rapid development of efficient vaccines led to the breakthrough of novel vaccine approaches such as mRNA-based vaccines saving millions of lives. Waning immunity of existing vaccines and emerging SARS-CoV-2 variants have required additional booster vaccinations and re-engineering of new versions of COVID-19 vaccines.
Collapse
|
6
|
Bronowicka-Szydełko A, Rabczyński M, Dumas I, Fiodorenko-Dumas Ż, Wojtczak B, Kotyra Ł, Kustrzeba-Wójcicka I, Lewandowski Ł, Ponikowska B, Kuzan A, Kluz J, Gamian A, Madziarska K. State of Knowledge About Thyroid Cancers in the Era of COVID-19-A Narrative Review. Biomedicines 2024; 12:2829. [PMID: 39767735 PMCID: PMC11672969 DOI: 10.3390/biomedicines12122829] [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: 10/26/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 01/03/2025] Open
Abstract
Thyroid cancer (TC), due to its heterogeneous nature, remains a clinical challenge. Many factors can initiate the carcinogenesis process of various types of TC, which complicates diagnosis and treatment. The presented review gathers current information on specific types of TC, taking into account the effects of the COVID-19 pandemic. It is likely that COVID-19 has influenced and continues to influence the function of the thyroid gland. A high percentage of patients with COVID-19 showing simultaneous pathological changes in the thyroid suggests that SARS-CoV-2 may disrupt the function of this gland and initiate pro-oxidative mechanisms, inflammatory states, and autoimmune diseases, thereby promoting the formation of neoplastic changes. Furthermore, changes in the expression of the ACE2, TMPRSS2, CLEC4M and DPP4 genes, observed in TC, also occur in COVID-19. Therefore, it is probable that the interaction of SARS-CoV-2 with thyroid cell receptors may initiate carcinogenesis in this gland. Additionally, some drugs used in TC therapy (e.g., levothyroxine) may increase the affinity of SARS-CoV-2 for cells, which could contribute to a more severe course of COVID-19 and the emergence of long-term symptoms (post-COVID-19). Moreover, the consequences of sanitary restrictions (limited access to medical services, reduction in endocrinological and oncological procedures) that took place in many countries during the COVID-19 pandemic may lead in the future to an increased number of missed diagnoses and the emergence of aggressive cancers.
Collapse
Affiliation(s)
- Agnieszka Bronowicka-Szydełko
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Maciej Rabczyński
- Clinical Department of Diabetology, Hypertension and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (M.R.); (J.K.); (K.M.)
| | - Ilias Dumas
- Department of Clinical Physiotherapy and Rehabilitation, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Żanna Fiodorenko-Dumas
- Department of Clinical Physiotherapy and Rehabilitation, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Beata Wojtczak
- University Center for General and Oncological Surgery, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Łukasz Kotyra
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Irena Kustrzeba-Wójcicka
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Łukasz Lewandowski
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Beata Ponikowska
- Department of Physiology and Pathophysiology, Division of Physiology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Aleksandra Kuzan
- Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Wroclaw University of Science and Technology, 51-377 Wroclaw, Poland;
| | - Joanna Kluz
- Clinical Department of Diabetology, Hypertension and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (M.R.); (J.K.); (K.M.)
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimantal Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Katarzyna Madziarska
- Clinical Department of Diabetology, Hypertension and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (M.R.); (J.K.); (K.M.)
| |
Collapse
|
7
|
Lee D, Cho M, Kim E, Seo Y, Cha JH. PD-L1: From cancer immunotherapy to therapeutic implications in multiple disorders. Mol Ther 2024; 32:4235-4255. [PMID: 39342430 PMCID: PMC11638837 DOI: 10.1016/j.ymthe.2024.09.026] [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: 02/25/2024] [Revised: 06/24/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
The PD-L1/PD-1 signaling pathway is the gold standard for cancer immunotherapy. Therapeutic antibodies targeting PD-1, such as nivolumab (Opdivo) and pembrolizumab (Keytruda), and PD-L1, including atezolizumab (Tecentriq), durvalumab (Imfinzi), and avelumab (Bavencio) have received Food and Drug Administration approval and are currently being used to treat various cancers. Traditionally, PD-L1 is known as an immune checkpoint protein that binds to the PD-1 receptor on its surface to inhibit the activity of T cells, which are the primary effector cells in antitumor immunity. However, it also plays a role in cancer progression, which goes beyond traditional understanding. Here, we highlight the multifaceted mechanisms of action of PD-L1 in cancer cell proliferation, transcriptional regulation, and systemic immune suppression. Moreover, we consider the potential role of PD-L1 in the development and pathogenesis of diseases other than cancer, explore PD-L1-focused therapeutic approaches for these diseases, and assess their clinical relevance. Through this review, we hope to provide deeper insights into the PD-L1/PD-1 signaling pathway and present a broad perspective on potential therapeutic approaches for cancer and other diseases.
Collapse
Affiliation(s)
- Daeun Lee
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Minjeong Cho
- Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea
| | - Eunseo Kim
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Youngbin Seo
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Jong-Ho Cha
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea; Biohybrid Systems Research Center, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
8
|
Subbarayan K, Al-Samadi A, Schäfer H, Massa C, Salo T, Biehl K, Vaxevanis CK, Ulagappan K, Wahbi W, Reimers M, Drexler F, Moreira-Soto A, Bachmann M, Seliger B. Altered ACE2 and interferon landscape in the COVID-19 microenvironment correlate with the anti-PD-1 response in solid tumors. Cell Mol Life Sci 2024; 81:473. [PMID: 39625479 PMCID: PMC11615173 DOI: 10.1007/s00018-024-05520-9] [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: 06/21/2024] [Revised: 10/03/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024]
Abstract
Angiotensensin-converting enzyme-2 (ACE2) is a receptor for SARS-CoV-2, allowing the virus to enter cells. Although tumor patients infected by SARS-CoV-2 often have a worse outcome, the expression, function and clinical relevance of ACE2 in tumors has not yet been thoroughly analyzed. In this study, RNA sequencing (RNA-seq) data from tumors, adjacent tissues and whole blood samples of COVID-19 patients from genome databases and from tumor cell lines and endothelial cells infected with different SARS-CoV-2 variants or transfected with an ACE2 expression vector (ACE2high) or mock (ACE2low) were analyzed for the expression of ACE2 and immune response relevant molecules in silico or by qPCR, flow cytometry, Western blot and/or RNA-seq. The differential expression profiles in ACE2high vs. ACE2low cells correlated with available SARS-CoV-2 RNA-seq datasets. ACE2high cells demonstrated upregulated mRNA and/or protein levels of HLA class I, programmed death ligand 1 (PD-L1), components of the antigen processing machinery (APM) and the interferon (IFN) signaling pathway compared to ACE2low cells. Co-cultures of ACE2high cells with peripheral blood mononuclear cells increased immune cell migration and infiltration towards ACE2high cells, apoptosis of ACE2high cells, release of innate immunity-related cytokines and altered NK cell-mediated cytotoxicity. Thus, ACE2 expression was associated in different model systems and upon SARS-CoV-2 infection with an altered host immunogenicity, which might influence the efficacy of immune checkpoint inhibitors. These results provide novel insights into the (patho)physiological role of ACE2 on immune response-relevant mechanisms and suggest an alternative strategy to reduce COVID-19 severity in infected tumor patients targeting the ACE2-induced IFN-PD-L1 axis.
Collapse
Affiliation(s)
- Karthikeyan Subbarayan
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Ahmed Al-Samadi
- Institute of Dentistry, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Joensuu, Finland
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Helene Schäfer
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Chiara Massa
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
- Institute of Translational Immunology, Brandenburg an der Havel, Germany
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, 90014, Finland
| | - Katharina Biehl
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Christoforos K Vaxevanis
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Kamatchi Ulagappan
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Matthias Reimers
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | | | | | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
- Institute of Translational Immunology, Brandenburg an der Havel, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
- Institute of Translational Medicine, Medical School Theodor Fontane, Hochstr. 29, 14770, Brandenburg an der Havel, Germany.
| |
Collapse
|
9
|
Almulla AF, Thipakorn Y, Zhou B, Vojdani A, Maes M. Immune activation and immune-associated neurotoxicity in Long-COVID: A systematic review and meta-analysis of 103 studies comprising 58 cytokines/chemokines/growth factors. Brain Behav Immun 2024; 122:75-94. [PMID: 39127088 DOI: 10.1016/j.bbi.2024.07.036] [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: 04/10/2024] [Revised: 07/18/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Multiple studies have shown that Long COVID (LC) disease is associated with heightened immune activation, as evidenced by elevated levels of inflammatory mediators. However, there is no comprehensive meta-analysis focusing on activation of the immune inflammatory response system (IRS) and the compensatory immunoregulatory system (CIRS) along with other immune phenotypes in LC patients. OBJECTIVES This meta-analysis is designed to explore the IRS and CIRS profiles in LC patients, the individual cytokines, chemokines, growth factors, along with C-reactive protein (CRP) and immune-associated neurotoxicity. METHODS To gather relevant studies for our research, we conducted a thorough search using databases such as PubMed, Google Scholar, and SciFinder, covering all available literature up to July 5th, 2024. RESULTS The current meta-analysis encompassed 103 studies that examined multiple immune profiles, C-reactive protein, and 58 cytokines/chemokines/growth factors in 5502 LC patients versus 5962 normal controls (NC). LC patients showed significant increases in IRS/CIRS ratio (standardized mean difference (SMD: 0.156, confidence interval (CI): 0.062;0.250), IRS (SMD: 0.338, CI: 0.236;0.440), M1 macrophage (SMD: 0.371, CI: 0.263;0.480), T helper (Th)1 (SMD: 0.316, CI: 0.185;0.446), Th17 (SMD: 0.439, CI: 0.302;0.577) and immune-associated neurotoxicity (SMD: 0.384, CI: 0.271;0.497). In addition, CRP and 21 different cytokines displayed significantly elevated levels in LC patients compared to NC. CONCLUSION LC disease is characterized by IRS activation and increased immune-associated neurotoxicity.
Collapse
Affiliation(s)
- Abbas F Almulla
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Yanin Thipakorn
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Bo Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China
| | - Aristo Vojdani
- Immunosciences Lab, Inc., Los Angeles, CA 90035, USA; Cyrex Laboratories, LLC, Phoenix, AZ 85034, USA
| | - Michael Maes
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Fitness and Biopsychological Technology Research Unit, Faculty of Medicine. Chulalongkorn University, Bangkok 10330, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria; Strategic Research and Innovation Program for the Development of MU - PLOVDIV-(SRIPD-MUP), European Union - NextGenerationEU; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, South Korea.
| |
Collapse
|
10
|
Liao K, Cheng J, Hu Y, Zhang B, Huang P, Liu J, Zhang W, Hu H, Bai X, Qian Y, Guo D, Ai K, Zhu Y, Huang L. Evaluation of the safety of PD-1/PD-L1 inhibitors for immunotherapy in patients with malignant tumors after COVID-19 infection: A single-center cohort study. Cancer Med 2024; 13:e70202. [PMID: 39377592 PMCID: PMC11459677 DOI: 10.1002/cam4.70202] [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: 03/12/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 10/09/2024] Open
Abstract
INTRODUCTION An increasing body of evidence suggests a close association between COVID-19 infection and the safety of PD-1/PD-L1 inhibitor therapy in cancer patients. However, the available data concerning these impacts remain limited and occasionally contradictory. MATERIAL AND METHODS We conducted a retrospective analysis of cancer patients who received PD-1/PD-L1 inhibitor therapy at the same institution from November 2022 to May 2023. After excluding patients with missing information, a total of 224 cases were included. In our study, immune-related adverse events (irAEs) that occurred during the hospitalization of patients were included in the analysis. Further analysis of inter-subgroup differences was conducted following a 1:2 propensity score matching. Statistical analyses were performed using the Fisher's exact, chi-squared, and Mann-Whitney U-tests. RESULT The results showed that no statistically significant differences between the two subgroups in the incidence of irAEs, changes in immune function before and after using PD-1/PD-L1 inhibitors, and alterations in hepatic and renal function (p > 0.05). CONCLUSION Our findings suggest that infection with COVID-19 does not significantly impact the safety of PD-1/PD-L1 inhibitors in cancer patients. Most cancer patients used PD-1/PD-L1 inhibitors during COVID-19 infection (asymptomatic or mild infection) did not experience exacerbation of their underlying condition, nor did they exhibit a substantial increase in toxic side effects.
Collapse
Affiliation(s)
- Kaili Liao
- Department of Clinical Laboratory, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Jinting Cheng
- School of Public Health, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yujie Hu
- The 1st Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Beining Zhang
- Queen Mary College, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Peng Huang
- Department of Oncology, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Jie Liu
- School of Public Health, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Wenyige Zhang
- Queen Mary College, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Huan Hu
- School of Public Health, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xinyi Bai
- School of Public Health, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yihui Qian
- The 2nd Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Daixin Guo
- School of Public Health, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Kun Ai
- Queen Mary College, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Yuchen Zhu
- The 1st Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Long Huang
- Department of Oncology, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
11
|
Calcaterra V, Loretelli C, Biganzoli D, Abdelsalam A, Marano G, Carelli S, Fiori L, Mannarino S, D'Auria E, Verduci E, De Santis R, Dilillo D, Fabiano V, Carlucci P, Maghraby E, Messa L, Cereda C, Fiorina P, Biganzoli E, Zuccotti G. Long-term cytokine profile in multisystem inflammatory disease among children. Cytokine 2024; 183:156744. [PMID: 39205361 DOI: 10.1016/j.cyto.2024.156744] [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: 03/10/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Multisystem inflammatory disease in children (MIS-C) is a post-infectious condition following coronavirus disease-19 infection. Long-term follow-up data suggests that initial clinical severity does not necessarily correlate with long-term outcomes. The long-term immunological response in children with MIS-C remains poorly understood. We analyzed cytokine profiles at diagnosis and during follow-up, in pediatric patients with MIS-C, exploring correlations among cytokine expressions and standard biochemical and hormonal test results. METHODS Twenty-five MIS-C patients (mean 9.4 ± 3.9) with complete test results at diagnosis and at 6- and 12-months follow-up were included in the study. Selected cytokines, such as IL-9, eotaxin, IP-10, MIP-1β, RANTES, MCP-1(MCAF), TNF-α, PDGF-B, IL-4, and MIP-1α, were included in the analysis. RESULTS IP-10, MCP-1 (MCAF), and MIP-1α levels normalized or nearly normalized at 6-12 months, the remaining cytokines, including IL-9, eotaxin, MIP-1β, RANTES, TNF-α, PDGF-B, IL-4, remained higher in MIS-C than in controls at our last follow-up time. At 6 months post-diagnosis, a mild negative correlation between triglycerides and HOMA-IR with MCP-1 (MCAF), IL-4, and Eotaxin was noted. At the 12-month follow-up we found a mild positive correlation of cortisol and ACTH levels with PDGF-B, MIP-1α, and TNF-α. Conversely, a negative correlation between these cytokines with fasting glucose and HOMA-IR was observed. CONCLUSIONS Our study findings highlight a notable cytokine-mediated inflammatory response in pediatric patients with MIS-C, characterized by sustained elevated levels over a 12-month monitoring period compared to the control group. We have identified various interrelationships among different cytokines, as well as correlations between heightened cytokine levels and metabolic and hormonal patterns. The pronounced inflammatory response underscores its involvement in acute organ damage, while its persistence suggests potential implications for long-term metabolic disorders.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy; Pediatric Department, Buzzi Children's Hospital, Milano, Italy
| | - Cristian Loretelli
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science, Università di Milano, Milano, Italy
| | - Davide Biganzoli
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milano, Italy
| | - Ahmed Abdelsalam
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science, Università di Milano, Milano, Italy
| | - Giuseppe Marano
- Medical Statistics Unit, Department of Biomedical and Clinical Sciences, "Luigi Sacco" University Hospital, University of Milano, Data Science Research Center, Milano, Italy
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milano, Italy
| | - Laura Fiori
- Pediatric Department, Buzzi Children's Hospital, Milano, Italy
| | | | - Enza D'Auria
- Pediatric Department, Buzzi Children's Hospital, Milano, Italy
| | - Elvira Verduci
- Pediatric Department, Buzzi Children's Hospital, Milano, Italy
| | | | - Dario Dilillo
- Pediatric Department, Buzzi Children's Hospital, Milano, Italy
| | - Valentina Fabiano
- Pediatric Department, Buzzi Children's Hospital, Milano, Italy; Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy.
| | | | - Erika Maghraby
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milano, Italy; Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy; Department of Biology and Biotechnology "L. Spallanzani" (DBB), University of Pavia, Pavia, Italy; Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milano, Italy
| | - Letizia Messa
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milano, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, Milano, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science, Università di Milano, Milano, Italy
| | - Elia Biganzoli
- Medical Statistics Unit, Department of Biomedical and Clinical Sciences, "Luigi Sacco" University Hospital, University of Milano, Data Science Research Center, Milano, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children's Hospital, Milano, Italy; Department of Biomedical and Clinical Sciences, University of Milano, Milano, Italy
| |
Collapse
|
12
|
Al-Aly Z, Davis H, McCorkell L, Soares L, Wulf-Hanson S, Iwasaki A, Topol EJ. Long COVID science, research and policy. Nat Med 2024; 30:2148-2164. [PMID: 39122965 DOI: 10.1038/s41591-024-03173-6] [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: 04/16/2024] [Accepted: 07/02/2024] [Indexed: 08/12/2024]
Abstract
Long COVID represents the constellation of post-acute and long-term health effects caused by SARS-CoV-2 infection; it is a complex, multisystem disorder that can affect nearly every organ system and can be severely disabling. The cumulative global incidence of long COVID is around 400 million individuals, which is estimated to have an annual economic impact of approximately $1 trillion-equivalent to about 1% of the global economy. Several mechanistic pathways are implicated in long COVID, including viral persistence, immune dysregulation, mitochondrial dysfunction, complement dysregulation, endothelial inflammation and microbiome dysbiosis. Long COVID can have devastating impacts on individual lives and, due to its complexity and prevalence, it also has major ramifications for health systems and economies, even threatening progress toward achieving the Sustainable Development Goals. Addressing the challenge of long COVID requires an ambitious and coordinated-but so far absent-global research and policy response strategy. In this interdisciplinary review, we provide a synthesis of the state of scientific evidence on long COVID, assess the impacts of long COVID on human health, health systems, the economy and global health metrics, and provide a forward-looking research and policy roadmap.
Collapse
Affiliation(s)
- Ziyad Al-Aly
- VA St. Louis Health Care System, Saint Louis, MO, USA.
- Washington University in St. Louis, Saint Louis, MO, USA.
| | - Hannah Davis
- Patient-led Research Collaborative, Calabasas, CA, USA
| | | | | | | | - Akiko Iwasaki
- Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Eric J Topol
- Scripps Institute, San Diego, California, CA, USA
| |
Collapse
|
13
|
Zheng HY, Song TZ, Zheng YT. Immunobiology of COVID-19: Mechanistic and therapeutic insights from animal models. Zool Res 2024; 45:747-766. [PMID: 38894519 PMCID: PMC11298684 DOI: 10.24272/j.issn.2095-8137.2024.062] [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: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 06/21/2024] Open
Abstract
The distribution of the immune system throughout the body complicates in vitro assessments of coronavirus disease 2019 (COVID-19) immunobiology, often resulting in a lack of reproducibility when extrapolated to the whole organism. Consequently, developing animal models is imperative for a comprehensive understanding of the pathology and immunology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This review summarizes current progress related to COVID-19 animal models, including non-human primates (NHPs), mice, and hamsters, with a focus on their roles in exploring the mechanisms of immunopathology, immune protection, and long-term effects of SARS-CoV-2 infection, as well as their application in immunoprevention and immunotherapy of SARS-CoV-2 infection. Differences among these animal models and their specific applications are also highlighted, as no single model can fully encapsulate all aspects of COVID-19. To effectively address the challenges posed by COVID-19, it is essential to select appropriate animal models that can accurately replicate both fatal and non-fatal infections with varying courses and severities. Optimizing animal model libraries and associated research tools is key to resolving the global COVID-19 pandemic, serving as a robust resource for future emerging infectious diseases.
Collapse
Affiliation(s)
- Hong-Yi Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Tian-Zhang Song
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China. E-mail:
| |
Collapse
|
14
|
Buendia‐Roldan I, Martínez‐Espinosa K, Aguirre M, Aguilar‐Duran H, Palma‐Lopez A, Palacios Y, Ruiz A, Ramón‐Luing LA, Ocaña‐Guzmán R, Pérez‐Rubio G, Falfán‐Valencia R, Selman M, Chavez‐Galan L. Persistence of lung structural and functional alterations at one year post-COVID-19 is associated with increased serum PD-L2 levels and altered CD4/CD8 ratio. Immun Inflamm Dis 2024; 12:e1305. [PMID: 39031504 PMCID: PMC11259001 DOI: 10.1002/iid3.1305] [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: 01/24/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Persistent respiratory symptoms and lung abnormalities post-COVID-19 are public health problems. This study evaluated biomarkers to stratify high-risk patients to the development or persistence of post-COVID-19 interstitial lung disease. METHODS One hundred eighteen patients discharged with residual lung abnormalities compatible with interstitial lung disease (COVID-ILD patients) after a severe COVID-19 were followed for 1 year (post-COVID-ILD patients). Physical examination, pulmonary function tests, and chest high-resolution computed tomography (HRCT) were performed. Soluble forms (s) of PD-L1, PD-L2, TIM-3, and GAL-9 were evaluated in serum and cell culture supernatant, as well as T-cells subsets and the transmembrane expression of PD-L1 and PD-L2 on the cell surface. RESULTS Eighty percent of the post-COVID-ILD patients normalized their lung function at 1-year follow-up, 8% presented COVID-independent ILD, and 12% still showed functional and HRCT alterations. PD-L2 levels were heterogeneous during acute COVID-19 (aCOVID); patients who increased (at least 30%) their sPD-L2 levels at 1 year post-COVID-19 and exhibited altered CD4/CD8 ratio showed persistence of chest tomographic and functional alterations. By contrast, patients who decreased sPD-L2 displayed a complete lung recovery. sPD-L1, sTIM-3, and sGAL-9 increased significantly during aCOVID and decreased in all patients after 1-year follow-up. CONCLUSION Increased sPD-L2 and an altered CD4/CD8 ratio after 12 months of aCOVID are associated with the persistence of lung lesions, suggesting that they may contribute to lung damage post-COVID-19.
Collapse
Affiliation(s)
- Ivette Buendia‐Roldan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | | | - Maria‐Jose Aguirre
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Hiram Aguilar‐Duran
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Alexia Palma‐Lopez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Yadira Palacios
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Andy Ruiz
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Lucero A. Ramón‐Luing
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Ranferi Ocaña‐Guzmán
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Gloria Pérez‐Rubio
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | | | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| | - Leslie Chavez‐Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío VillegasMexico CityMexico
| |
Collapse
|
15
|
Fracella M, Mancino E, Nenna R, Virgillito C, Frasca F, D'Auria A, Sorrentino L, Petrarca L, La Regina D, Matera L, Di Mattia G, Caputo B, Antonelli G, Pierangeli A, Viscidi RP, Midulla F, Scagnolari C. Age-related transcript changes in type I interferon signaling in children and adolescents with long COVID. Eur J Immunol 2024; 54:e2350682. [PMID: 38522030 DOI: 10.1002/eji.202350682] [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: 07/25/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/25/2024]
Abstract
SARS-CoV-2 typically causes mild symptoms in children, but evidence suggests that persistent immunopathological changes may lead to long COVID (LC). To explore the interplay between LC and innate immunity, we assessed the type I interferon (IFN-I) response in children and adolescents with LC symptoms (LC; n = 28). This was compared with age-matched SARS-CoV-2 recovered participants without LC symptoms (MC; n = 28) and healthy controls (HC; n = 18). We measured the mRNA expression of IFN-I (IFN-α/β/ε/ω), IFN-I receptor (IFNAR1/2), and ISGs (ISG15, ISG56, MxA, IFI27, BST2, LY6E, OAS1, OAS2, OAS3, and MDA5) in PBMCs collected 3-6 months after COVID-19. LC adolescents (12-17 years) had higher transcript levels of IFN-β, IFN-ε, and IFN-ω than HC, whereas LC children (6-11 years) had lower levels than HC. In adolescents, increased levels of IFN-α, IFN-β, and IFN-ω mRNAs were found in the LC group compared with MC, while lower levels were observed in LC children than MC. Adolescents with neurological symptoms had higher IFN-α/β mRNA levels than MC. LC and MC participants showed decreased expression of ISGs and IFNAR1, but increased expression of IFNAR2, than HC. Our results show age-related changes in the expression of transcripts involved in the IFN-I signaling pathway in children and adolescents with LC.
Collapse
Affiliation(s)
- Matteo Fracella
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Enrica Mancino
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Raffaella Nenna
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Chiara Virgillito
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Federica Frasca
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessandra D'Auria
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Leonardo Sorrentino
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Laura Petrarca
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Domenico La Regina
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Luigi Matera
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Greta Di Mattia
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Beniamino Caputo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Guido Antonelli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Alessandra Pierangeli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Raphael P Viscidi
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fabio Midulla
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
Bonora BM, Marassi M, Fogar P, Zuin J, Cappellari R, Marinello S, Ferrari A, Cattelan A, Avogaro A, Basso D, Fadini GP. Circulating haematopoietic stem cells and long-term outcomes of COVID-19. Eur J Clin Invest 2024; 54:e14150. [PMID: 38088242 DOI: 10.1111/eci.14150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND AIMS An acute depletion of circulating haematopoietic stem/progenitor cells (HSPCs) occurs during COVID-19, especially among patients with a poorer disease course. We herein examined whether HSPCs levels at hospital admission for COVID-19 predict 1-year mortality and the long-COVID syndrome. MATERIALS AND METHODS Patients hospitalized for COVID-19 in an infectious disease ward were consecutively enrolled. Circulating HSPC levels were assessed by flow cytometry as cells expressing CD34 and/or CD133. Follow-up was performed for 12 months after hospitalization through the review of electronic medical records and demographic local registers. RESULTS The study included 100 patients, 36 of whom reported symptoms of long-COVID and 20 died during follow-up. The reduction of 1-SD of HSPCs was associated with a 3- to 5-fold increase in the risk of 1-year mortality. Age, admission hyperglycaemia, C-reactive protein peak, liver enzymes, the need of high-flow oxygen and/or invasive ventilation were predictors of mortality at univariate analysis. Among pre-existing comorbidities, coronary heart disease and chronic kidney disease, but not diabetes, were associated with 1-year mortality. In multivariate analyses, HSPCs remained significantly associated with 1-year mortality independently of confounders. The development of pneumonia an in-hospital treatment with glucocorticoids and convalescent plasma were associated with long-COVID symptoms at follow-up. HSPCs, diabetes and other comorbidities were not predictors of long-COVID. CONCLUSIONS In a cohort of patients hospitalized for COVID-19, lower HSPC levels at the time of admission were independent predictors of 1-year mortality. However, COVID-19 severity, but not HSPC level, was significantly associated with the development of long-COVID symptoms.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University Hospital of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Marella Marassi
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Paola Fogar
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Jenny Zuin
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | | | - Serena Marinello
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Anna Ferrari
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Daniela Basso
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University Hospital of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
17
|
Liu H, Guo N, Zheng Q, Zhang Q, Chen J, Cai Y, Luo Q, Xu Q, Chen X, Yang S, Zhang S. Association of interleukin-6, ferritin, and lactate dehydrogenase with venous thromboembolism in COVID-19: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:324. [PMID: 38493138 PMCID: PMC10943892 DOI: 10.1186/s12879-024-09205-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/07/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is frequntly accompanied by venous thromboembolism (VTE), and its mechanism may be related to the abnormal inflammation and immune status of COVID-19 patients. It has been proved that interleukin-6 (IL-6), ferritin and lactate dehydrogenase (LDH) may play an important role in the occurrence of VTE in COVID-19 infection. But whether they can server as predictors for VTE in COVID-19 is still unclear. In this study, we performed a systematic review and meta-analysis to compare IL-6, ferritin and LDH in VTE and non-VTE COVID-19 patients in order to shed light on the prevention and treatment of VTE. METHODS Related literatures were searched in PubMed, Embase, Web of Science, Google Scholar, China National Knowledge Infrastructure (CNKI), WANGFANG. COVID-19 patients were divided into VTE group and non-VTE group. Meta-analysis was then conducted to compare levels of IL-6, ferritin and LDH between the two groups. RESULTS We finally included and analyzed 17 literatures from January 2019 to October 2022. There was a total of 7,035 COVID-19 patients, with a weighted mean age of 60.01 years. Males accounted for 62.64% and 61.34% patients were in intensive care unit (ICU). Weighted mean difference (WMD) of IL-6, ferritin and LDH was 31.15 (95% CI: 9.82, 52.49), 257.02 (95% CI: 51.70, 462.33) and 41.79 (95% CI: -19.38, 102.96), respectively. The above results indicated that than compared with non-VTE group, VTE group had significantly higher levels of IL-6 and ferritin but similar LDH. CONCLUSION This systematic review and meta-analysis pointed out that elevated levels of IL-6 and ferritin were significantly possitive associated with VTE, thus could be used as biological predictive indicators of VTE among COVID-19 patients. However, no association was found between level of LDH and VTE. Therefore, close monitoring of changes in IL-6 and ferritin concentrations is of great value in assisting clinicans to rapidly identify thrombotic complications among COVID-19 patients, hence facilitating the timely effective managment. Further studies are required in terms of the clinical role of cytokines in the occurrence of VTE among COVID-19 infection, with more reliable systematic controls and interventional trials.
Collapse
Affiliation(s)
- Haiyu Liu
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Ningjing Guo
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Qixian Zheng
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Qianyuan Zhang
- Department of General Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Jinghan Chen
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Yuanyuan Cai
- Department of General Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Qiong Luo
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Qian Xu
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China
| | - Xiangqi Chen
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China.
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Fuzhou, Fujian, 350001, P.R. China.
| | - Sheng Yang
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China.
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Fuzhou, Fujian, 350001, P.R. China.
| | - Suyun Zhang
- Department of Internal Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, P.R. China.
| |
Collapse
|
18
|
Chambers CD, Song J, da Silva Antunes R, Sette A, Franco A. T Cell Responses in Pregnant Women Who Received mRNA-Based Vaccination to Prevent COVID-19 Revealed Unknown Exposure to the Natural Infection and Numerous SARS-CoV-2-Specific CD4- CD8- Double Negative T Cells and Regulatory T Cells. Int J Mol Sci 2024; 25:2031. [PMID: 38396707 PMCID: PMC10889590 DOI: 10.3390/ijms25042031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
We studied T-cell responses to SARS-CoV-2 in 19 pregnant subjects at different gestational weeks who received three doses of mRNA-based vaccination to prevent COVID-19. SARS-CoV-2 peptide pools were used for T-cell recognition studies: peptides were 15 amino acids long and had previously been defined in COVID-19-convalescent subjects. T-cell activation was evaluated with the AIM assay. Most subjects showed coordinated, spike-specific CD4+ and CD8+ T-cell responses and the development of T cell memory. Non-spike-specific T cells in subjects who were not aware of previous COVID-19 infection suggested a prior undetected, asymptomatic infection. CD4- CD8- double negative (DN) T cells were numerous, of which a percentage was specific for SARS-CoV-2 spike peptides. Regulatory T cells (Treg), both spike- and non-spike-specific, were also greatly expanded. Two Treg populations were defined: a population differentiated from naïve T cells, and pTreg, reverting from pro-inflammatory T cells. The Treg cells expressed CCR6, suggesting homing to the endometrium and vaginal epithelial cells. The pregnant women responded to SARS-CoV-2 vaccination. Asymptomatic COVID-19 was revealed by the T cell response to the non-spike peptides. The numerous DN T cells and Treg pointed our attention to new aspects of the adaptive immune response in vaccine recipients.
Collapse
Affiliation(s)
- Christina D. Chambers
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (C.D.C.); (J.S.)
| | - Jaeyoon Song
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (C.D.C.); (J.S.)
| | - Ricardo da Silva Antunes
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA (A.S.)
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA 92093, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA (A.S.)
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA 92093, USA
| | - Alessandra Franco
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (C.D.C.); (J.S.)
| |
Collapse
|
19
|
Curran CS, Cui X, Li Y, Jeakle M, Sun J, Demirkale CY, Minkove S, Hoffmann V, Dhamapurkar R, Chumbris S, Bolyard C, Iheanacho A, Eichacker PQ, Torabi-Parizi P. Anti-PD-L1 therapy altered inflammation but not survival in a lethal murine hepatitis virus-1 pneumonia model. Front Immunol 2024; 14:1308358. [PMID: 38259435 PMCID: PMC10801642 DOI: 10.3389/fimmu.2023.1308358] [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: 10/06/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Because prior immune checkpoint inhibitor (ICI) therapy in cancer patients presenting with COVID-19 may affect outcomes, we investigated the beta-coronavirus, murine hepatitis virus (MHV)-1, in a lethal pneumonia model in the absence (Study 1) or presence of prior programmed cell death ligand-1 (PD-L1) antibody (PD-L1mAb) treatment (Study 2). Methods In Study 1, animals were inoculated intratracheally with MHV-1 or vehicle and evaluated at day 2, 5, and 10 after infection. In Study 2, uninfected or MHV-1-infected animals were pretreated intraperitoneally with control or PD-L1-blocking antibodies (PD-L1mAb) and evaluated at day 2 and 5 after infection. Each study examined survival, physiologic and histologic parameters, viral titers, lung immunophenotypes, and mediator production. Results Study 1 results recapitulated the pathogenesis of COVID-19 and revealed increased cell surface expression of checkpoint molecules (PD-L1, PD-1), higher expression of the immune activation marker angiotensin converting enzyme (ACE), but reduced detection of the MHV-1 receptor CD66a on immune cells in the lung, liver, and spleen. In addition to reduced detection of PD-L1 on all immune cells assayed, PD-L1 blockade was associated with increased cell surface expression of PD-1 and ACE, decreased cell surface detection of CD66a, and improved oxygen saturation despite reduced blood glucose levels and increased signs of tissue hypoxia. In the lung, PD-L1mAb promoted S100A9 but inhibited ACE2 production concomitantly with pAKT activation and reduced FOXO1 levels. PD-L1mAb promoted interferon-γ but inhibited IL-5 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production, contributing to reduced bronchoalveolar lavage levels of eosinophils and neutrophils. In the liver, PD-L1mAb increased viral clearance in association with increased macrophage and lymphocyte recruitment and liver injury. PD-L1mAb increased the production of virally induced mediators of injury, angiogenesis, and neuronal activity that may play role in COVID-19 and ICI-related neurotoxicity. PD-L1mAb did not affect survival in this murine model. Discussion In Study 1 and Study 2, ACE was upregulated and CD66a and ACE2 were downregulated by either MHV-1 or PD-L1mAb. CD66a is not only the MHV-1 receptor but also an identified immune checkpoint and a negative regulator of ACE. Crosstalk between CD66a and PD-L1 or ACE/ACE2 may provide insight into ICI therapies. These networks may also play role in the increased production of S100A9 and neurological mediators in response to MHV-1 and/or PD-L1mAb, which warrant further study. Overall, these findings support observational data suggesting that prior ICI treatment does not alter survival in patients presenting with COVID-19.
Collapse
Affiliation(s)
- Colleen S. Curran
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Mark Jeakle
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Cumhur Y. Demirkale
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Samuel Minkove
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Victoria Hoffmann
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, United States
| | - Rhea Dhamapurkar
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Symya Chumbris
- Texcell North-America, Inc., Frederick, MD, United States
| | | | | | - Peter Q. Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Parizad Torabi-Parizi
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
20
|
Davarpanah MA, Adatorwovor R, Mansoori Y, Ramsheh FSR, Parsa A, Hajiani M, Faramarzi H, Kavuluru R, Asadipooya K. Combination of spironolactone and sitagliptin improves clinical outcomes of outpatients with COVID-19: a prospective cohort study. J Endocrinol Invest 2024; 47:235-243. [PMID: 37354247 DOI: 10.1007/s40618-023-02141-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND There are evidences showing that sitagliptin and spironolactone can potentially improve the clinical outcomes of COVID-19 cases. In this observational study on acutely symptomatic outpatient COVID-19 cases, we investigated the effects of spironolactone and sitagliptin on the outcomes of the disease. METHODS This is a prospective, naturally randomized cohort study. We followed mild to moderate symptomatic COVID-19 patients, who were treated with either combination (spironolactone 100 mg daily and sitagliptin 100 mg daily) or standard (steroid, antiviral and/or supportive care) therapy up to 30 days. The primary outcome was hospitalization rate. The secondary outcomes included ER visit, duration of disease, and complications, such as hypoglycemia, low blood pressure or altered mental status. RESULTS Of the 206 patients referred to clinics randomly, 103 received standard therapy and 103 treated with combination therapy. There were no significant differences in baseline characteristics, except for slightly higher clinical score in control group (6.92 ± 4.01 control, 4.87 ± 2.92 combination; P < 0.0001). Treatment with combination therapy was associated with lower admission rate (5.8% combination, 22.3% control; P = 0.0011), ER visits (7.8% combination, 23.3% control; P = 0.0021) and average duration of symptoms (6.67 ± 2.30 days combination, 18.71 ± 6.49 days control; P ≤ 0.0001). CONCLUSIONS The combination of sitagliptin and spironolactone reduced duration of COVID infection and hospital visits better than standard therapeutic approaches in outpatients with COVID-19. The effects of combination of sitagliptin and spironolactone in COVID-19 patients should be further verified in a double-blind, randomized, placebo-controlled trial.
Collapse
Affiliation(s)
- M A Davarpanah
- Shiraz HIV/AIDS Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - R Adatorwovor
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Y Mansoori
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - F S R Ramsheh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - A Parsa
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - M Hajiani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - H Faramarzi
- Department of Community Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - R Kavuluru
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - K Asadipooya
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Barnstable Brown Diabetes and Obesity Center, University of Kentucky, 2195 Harrodsburg Rd, Suite 125, Lexington, KY, 40504, USA.
| |
Collapse
|
21
|
Liu X, Xiong W, Ye M, Lu T, Yuan K, Chang S, Han Y, Wang Y, Lu L, Bao Y. Non-coding RNAs expression in SARS-CoV-2 infection: pathogenesis, clinical significance, and therapeutic targets. Signal Transduct Target Ther 2023; 8:441. [PMID: 38057315 PMCID: PMC10700414 DOI: 10.1038/s41392-023-01669-0] [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: 12/09/2022] [Revised: 09/12/2023] [Accepted: 09/28/2023] [Indexed: 12/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has been looming globally for three years, yet the diagnostic and treatment methods for COVID-19 are still undergoing extensive exploration, which holds paramount importance in mitigating future epidemics. Host non-coding RNAs (ncRNAs) display aberrations in the context of COVID-19. Specifically, microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) exhibit a close association with viral infection and disease progression. In this comprehensive review, an overview was presented of the expression profiles of host ncRNAs following SARS-CoV-2 invasion and of the potential functions in COVID-19 development, encompassing viral invasion, replication, immune response, and multiorgan deficits which include respiratory system, cardiac system, central nervous system, peripheral nervous system as well as long COVID. Furthermore, we provide an overview of several promising host ncRNA biomarkers for diverse clinical scenarios related to COVID-19, such as stratification biomarkers, prognostic biomarkers, and predictive biomarkers for treatment response. In addition, we also discuss the therapeutic potential of ncRNAs for COVID-19, presenting ncRNA-based strategies to facilitate the development of novel treatments. Through an in-depth analysis of the interplay between ncRNA and COVID-19 combined with our bioinformatic analysis, we hope to offer valuable insights into the stratification, prognosis, and treatment of COVID-19.
Collapse
Affiliation(s)
- Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Wandi Xiong
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, 570228, Haikou, China
| | - Maosen Ye
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Suhua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Yongxiang Wang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117, Jinan, Shandong, China.
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China.
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China.
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117, Jinan, Shandong, China.
- School of Public Health, Peking University, 100191, Beijing, China.
| |
Collapse
|
22
|
Kaiho T, Suzuki H, Hata A, Matsumoto H, Tanaka K, Sakairi Y, Motohashi S, Yoshino I. Targeting PD-1/PD-L1 inhibits rejection in a heterotopic tracheal allograft model of lung transplantation. Front Pharmacol 2023; 14:1298085. [PMID: 38026994 PMCID: PMC10657857 DOI: 10.3389/fphar.2023.1298085] [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: 09/21/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Immune checkpoint molecules such as programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1) have revolutionized the field of lung cancer treatment. As part of our study, we examined the role of these proteins in acute rejection in a mouse model of heterotopic tracheal transplantation. Recipient mice were untreated (Allo group) or treated with anti-PD-L1 (aPDL1 group) or PD-L1 Fc recombinant protein (PD-L1 Fc group). A further group of C57BL/6 mice received isografts (Iso group). The occlusion rate was significantly higher in the Allo group than in the Iso group (p = 0.0075), and also higher in the aPD-L1 group (p = 0.0066) and lower in the PD-L1 Fc group (p = 0.030) than in the Allo group. PD-L1 Fc recombinant protein treatment significantly decreased interleukin-6 and interferon-γ levels and reduced the CD4+/CD8+ T cell ratio, without increasing PD-1 and T-cell immunoglobulin mucin 3 expression in CD4+ T cells. These data suggest that PD-L1 Fc recombinant protein decreases the levels of inflammatory cytokines and the proportion of CD4+ T cells without exhaustion. The PD-L1-mediated immune checkpoint mechanism was associated with rejection in the murine tracheal transplant model, suggesting a potential novel target for immunotherapy in lung transplantation.
Collapse
Affiliation(s)
- Taisuke Kaiho
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidemi Suzuki
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Hata
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hiroki Matsumoto
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kazuhisa Tanaka
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yuichi Sakairi
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinichiro Motohashi
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Medical Immunology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
23
|
Karavanaki K, Karanasios S, Soldatou A, Tsolia M. SARS-CoV-2 vaccination in children and adolescents with and without type 1 diabetes mellitus. Endocrine 2023; 82:226-236. [PMID: 37587391 PMCID: PMC10543146 DOI: 10.1007/s12020-023-03471-y] [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: 04/19/2023] [Accepted: 07/27/2023] [Indexed: 08/18/2023]
Abstract
Adults with Diabetes Mellitus (DM) have increased risk of severe clinical presentation during COVID-19 infection, while children and adolescents with type 1 diabetes (T1D) have the same mild clinical course as their healthy peers, especially those with optimal glycemic control. The present review focuses on the necessity of COVID-19 vaccination among children and adolescents with T1D, and also in their non-diabetic peers. The efficacy and safety of COVID-19 vaccines are also discussed, as well as their various side-effects, ranging from common mild to very rare and serious ones. Furthermore, the results of COVID-19 vaccination of adolescents with and without T1D are reported, as well as the efficacy and concerns about childhood vaccination. It is concluded that patients with DM of all age groups should maintain optimal diabetic control in order to avoid glycemic deterioration during COVID-19 infection. Furthermore, despite the very rare and serious complications of COVID-19 vaccines, vaccination against COVID-19 is recommended for children and adolescents with T1D to prevent glycemic deterioration and rare but serious complications of COVID-19 infection.
Collapse
Affiliation(s)
- Kyriaki Karavanaki
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece
| | - Spyridon Karanasios
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece
| | - Alexandra Soldatou
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece.
| | - Maria Tsolia
- Diabetes and Metabolism Clinic, 2nd Department of Pediatrics, National and Kapodistrian University of Athens, "P&A Kyriakou" Children's Hospital, Athens, Greece
| |
Collapse
|
24
|
Zhang P, Wang Y, Miao Q, Chen Y. The therapeutic potential of PD-1/PD-L1 pathway on immune-related diseases: Based on the innate and adaptive immune components. Biomed Pharmacother 2023; 167:115569. [PMID: 37769390 DOI: 10.1016/j.biopha.2023.115569] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023] Open
Abstract
Currently, immunotherapy targeting programmed cell death 1 (PD-1) or programmed death ligand 1 (PD-L1) has revolutionized the treatment strategy of human cancer patients. Meanwhile, PD-1/PD-L1 pathway has also been implicated in the pathogenesis of many immune-related diseases, such as autoimmune diseases, chronic infection diseases and adverse pregnancy outcomes, by regulating components of the innate and adaptive immune systems. Given the power of the new therapy, a better understanding of the regulatory effects of PD-1/PD-L1 pathway on innate and adaptive immune responses in immune-related diseases will facilitate the discovery of novel biomarkers and therapeutic drug targets. Targeting this pathway may successfully halt or potentially even reverse these pathological processes. In this review, we discuss recent major advances in PD-1/PD-L1 axis regulating innate and adaptive immune components in immune-related diseases. We reveal that the impact of PD-1/PD-L1 axis on the immune system is complex and manifold and multi-strategies on the targeted PD-1/PD-L1 axis are taken in the treatment of immune-related diseases. Consequently, targeting PD-1/PD-L1 pathway, alone or in combination with other treatments, may represent a novel strategy for future therapeutic intervention on immune-related diseases.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yuting Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Qianru Miao
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Ying Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
25
|
Roe K. Pathogen regulatory RNA usage enables chronic infections, T-cell exhaustion and accelerated T-cell exhaustion. Mol Cell Biochem 2023; 478:2505-2516. [PMID: 36941498 PMCID: PMC10027582 DOI: 10.1007/s11010-023-04680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 02/15/2023] [Indexed: 03/23/2023]
Abstract
Pathogens evade or disable cellular immune defenses using regulatory ribonucleic acids (RNAs), including microRNAs and long non-coding RNAs. Pathogenic usage of regulatory RNA enables chronic infections. Chronic infections, using host regulatory RNAs and/or creating pathogenic regulatory RNAs against cellular defenses, can cause T-cell exhaustion and latent pathogen reactivations. Concurrent pathogen infections of cells enable several possibilities. A first pathogen can cause an accelerated T-cell exhaustion for a second pathogen cellular infection. Accelerated T-cell exhaustion for the second pathogen weakens T-cell targeting of the second pathogen and enables a first-time infection by the second pathogen to replicate quickly and extensively. This can induce a large antibody population, which may be inadequately targeted against the second pathogen. Accelerated T-cell exhaustion can explain the relatively short median and average times from diagnosis to mortality in some viral epidemics, e.g., COVID-19, where the second pathogen can lethally overwhelm individuals' immune defenses. Alternatively, if an individual survives, the second pathogen could induce a very high titer of antigen-antibody immune complexes. If the antigen-antibody immune complex titer quickly becomes very high, it can exceed the immune system's phagocytic capability in immuno-deficient individuals, resulting in a Type III hypersensitivity immune reaction. Accelerated T-cell exhaustion in immuno-deficient individuals can be a fundamental cause of several hyperinflammatory diseases and autoimmune diseases. This would be possible when impaired follicular helper CD4+ T-cell assistance to germinal center B-cell somatic hypermutation, affinity maturation and isotype switching of antibodies results in high titers of inadequate antibodies, and this initiates a Type III hypersensitivity immune reaction with proteinase releases which express or expose autoantigens.
Collapse
|
26
|
Sabbatino F, Pagliano P, Sellitto C, Stefanelli B, Corbi G, Manzo V, De Bellis E, Liguori L, Salzano FA, Pepe S, Filippelli A, Conti V. Different Prognostic Role of Soluble PD-L1 in the Course of Severe and Non-Severe COVID-19. J Clin Med 2023; 12:6812. [PMID: 37959277 PMCID: PMC10649852 DOI: 10.3390/jcm12216812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Understanding the link between COVID-19 and patient immune characteristics is crucial. We previously demonstrated that high levels of the soluble Programmed Death-Ligand1 (sPD-L1) at the beginning of the infection correlated with low lymphocyte number and high C-reactive protein (CRP), longer length of stay (LOS), and death. This study investigated whether sPD-L1 can be a prognosis biomarker during COVID-19. Severe and non-severe COVID-19 patients were enrolled at the University Hospital of Salerno. During hospitalization, at admission, and after 12-14 days, patients' data were collected, and sPD-L1 levels were measured by enzyme-linked immunosorbent assay. The peripheral lymphocyte number negatively correlated with the time of negativization (p = 0.006), length of stay (LOS) (p = 0.032), and CRP (p = 0.004), while sPD-L1 positively correlated with LOS (p = 0.015). Patients with increased sPD-L1 and lymphocyte number showed a shorter LOS than those with decreased sPD-L1 and lymphocyte number (p = 0.038) and those with increased sPD-L1 and decreased lymphocyte number (p = 0.025). Moreover, patients with increased sPD-L1 and decreased CRP had a shorter LOS than those with increased sPD-L1 and CRP (p = 0.034) and those with decreased sPD-L1 and CRP (p = 0.048). In conclusion, while at an early phase of COVID-19, sPD-L1 promotes an immune escape, later, it might act to dampen an excessive immune response, proving its role in COVID-19 prognosis.
Collapse
Affiliation(s)
- Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Oncology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, 84131 Salerno, Italy
| | - Pasquale Pagliano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Infectious Disease Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, 84131 Salerno, Italy
| | - Carmine Sellitto
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Clinical Pharmacology Unit, San Giovanni di Dio e Ruggi d’Aragona University Hospital, Via San Leonardo 1, 84131 Salerno, Italy
| | - Berenice Stefanelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
| | - Graziamaria Corbi
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy
| | - Valentina Manzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Clinical Pharmacology Unit, San Giovanni di Dio e Ruggi d’Aragona University Hospital, Via San Leonardo 1, 84131 Salerno, Italy
| | - Emanuela De Bellis
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
| | - Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Francesco Antonio Salzano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Otolaryngology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, 84131 Salerno, Italy
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Oncology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, 84131 Salerno, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Clinical Pharmacology Unit, San Giovanni di Dio e Ruggi d’Aragona University Hospital, Via San Leonardo 1, 84131 Salerno, Italy
| | - Valeria Conti
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.S.); (P.P.); (C.S.); (B.S.); (V.M.); (E.D.B.); (F.A.S.); (S.P.); (A.F.); (V.C.)
- Clinical Pharmacology Unit, San Giovanni di Dio e Ruggi d’Aragona University Hospital, Via San Leonardo 1, 84131 Salerno, Italy
| |
Collapse
|
27
|
Silva BSDA, Pereira T, Minuzzi LG, Padilha CS, Figueiredo C, Olean-Oliveira T, dos Santos IVM, von Ah Morano AE, Marchioto Júnior O, Ribeiro JPJ, Dos Santos VR, Seelaender M, Teixeira AA, Dos Santos RVT, Lemos VDA, Freire APCF, Dorneles GP, Marmett B, Olean-Oliveira A, Teixeira MFS, Seraphim PM, Caseiro A, Pinho RA, Islam H, Little JP, Krüger K, Rosa-Neto JC, Coelho-E-Silva MJ, Lira FS. Mild to moderate post-COVID-19 alters markers of lymphocyte activation, exhaustion, and immunometabolic responses that can be partially associated by physical activity level- an observational sub-analysis fit- COVID study. Front Immunol 2023; 14:1212745. [PMID: 37753077 PMCID: PMC10518618 DOI: 10.3389/fimmu.2023.1212745] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/10/2023] [Indexed: 09/28/2023] Open
Abstract
Aim This study aimed to evaluate if physical activity is associated with systemic and cellular immunometabolic responses, in young adults after mild-to-moderate COVID-19 infection. Methods Mild- to- moderate post-COVID-19 patients (70.50 ± 43.10 days of diagnosis; age: 29.4 (21.9- 34.9) years; BMI: 25.5 ± 4.3 kg m2 n = 20) and healthy age-matched controls (age: 29.3 (21.2 - 32.6) years; BMI: 25.4 ± 4.7 kg m2; n = 20) were evaluated. Physical activity levels (PAL), body composition, dietary habits, muscular and pulmonary function, mental health, sleep quality, metabolic parameters, immune phenotypic characterization, stimulated whole blood and PBMC culture (cytokine production), mRNA, and mitochondrial respiration in PBMCs were evaluated. Results The post-COVID-19 group exhibited lower levels of moderate to vigorous physical activity (MVPA) (p = 0.038); therefore, all study comparisons were performed with adjustment for MVPA. Post-COVID-19 impacted the pulmonary function (FEV1, FEV1%pred, FVC, and FVC %pred) compared with the control (p adjusted by MVPA (p adj) <0.05). Post-COVID-19 exhibited lower levels of serum IL-6 (p adj <0.01), whereas it showed higher serum IL-10, triglyceride, leptin, IgG, ACE activity, TNFRSF1A, and PGE2 (p adj <0.05) levels compared with controls. Post-COVID-19 presented a lower percentage of Treg cells (p adj = 0.03) and altered markers of lymphocyte activation and exhaustion (lower CD28 expression in CD8+ T cells (p adj = 0.014), whereas CD4+T cells showed higher PD1 expression (p adj = 0.037)) compared with the control group. Finally, post- COVID-19 presented an increased LPS-stimulated whole- blood IL-10 concentration (p adj <0.01). When exploring mitochondrial respiration and gene expression in PBMCs, we observed a higher LEAK state value (p adj <0.01), lower OXPHOS activity (complex I) (p adj = 0.04), and expression of the Rev-Erb-α clock mRNA after LPS stimulation in the post-COVID-19 patients than in the control (p adj <0.01). Mainly, PAL was associated with changes in IL-10, triglyceride, and leptin levels in the plasma of post-COVID-19 patients. PAL was also associated with modulation of the peripheral frequency of Treg cells and the expression of PD-1 in CD8+ T cells, although it abrogated the statistical effect in the analysis of TNF-α and IL-6 production by LPS- and PMA-stimulated PBMC of post-COVID-19 patients. Conclusion Young adults after mild-to-moderate SARS-CoV-2 infection appeared to have lower physical activity levels, which can be associated with clinical and immunometabolic responses in a complex manner.
Collapse
Affiliation(s)
- Bruna Spolador de Alencar Silva
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Telmo Pereira
- Polytechnic Institute of Coimbra, Coimbra Health School, Coimbra, Portugal
| | - Luciele Guerra Minuzzi
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Camila Souza Padilha
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Caique Figueiredo
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Tiago Olean-Oliveira
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Ivete Vera Medeiros dos Santos
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Ana Elisa von Ah Morano
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Osmar Marchioto Júnior
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - José Procópio Jabur Ribeiro
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Vanessa Ribeiro Dos Santos
- Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Marília Seelaender
- Cancer Metabolism Research Group, LIM26-HC, FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | - Ana Paula Coelho Figueira Freire
- Department of Health Sciences, Central Washington University, Ellensburg, WA, United States
- Physiotherapy Department, Universidade do Oeste Paulista (UNOESTE), Presidente Prudente, Brazil
| | - Gilson Pires Dorneles
- Laboratory of Cellular and Molecular Immunology, Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Bruna Marmett
- Laboratory of Cellular and Molecular Immunology, Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - André Olean-Oliveira
- Department of Chemistry and Biochemistry, School of Science and Technology, Universidade Estadual Paulista (UNESP), Presidente Prudente, SP, Brazil
| | - Marcos F. S. Teixeira
- Department of Chemistry and Biochemistry, School of Science and Technology, Universidade Estadual Paulista (UNESP), Presidente Prudente, SP, Brazil
| | - Patrícia M. Seraphim
- Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Armando Caseiro
- Polytechnic Institute of Coimbra, Coimbra Health School, Coimbra, Portugal
| | - Ricardo Aurino Pinho
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Parana, Curitiba, Brazil
| | - Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Jonathan Peter Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - José César Rosa-Neto
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Manuel-João Coelho-E-Silva
- Faculty of Sport Sciences and Physical Education, Research Center for Sport and Physical Activity (uid/dtp/04213/2020), Universidade de Coimbra, Coimbra, Portugal
| | - Fábio Santos Lira
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
- Faculty of Sport Sciences and Physical Education, Research Center for Sport and Physical Activity (uid/dtp/04213/2020), Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
28
|
Pongkunakorn T, Manosan T, Surawit A, Ophakas S, Mongkolsucharitkul P, Pumeiam S, Suta S, Pinsawas B, Sookrung N, Saelim N, Mahasongkram K, Prangtaworn P, Tungtrongchitr A, Tangjittipokin W, Mangmee S, Boonnak K, Narkdontri T, Teerawattanapong N, Wanitphadeedecha R, Mayurasakorn K. Immune Response after SARS-CoV-2 Infection with Residual Post-COVID Symptoms. Vaccines (Basel) 2023; 11:1413. [PMID: 37766091 PMCID: PMC10535557 DOI: 10.3390/vaccines11091413] [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: 07/19/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Many patients develop post-acute COVID syndrome (long COVID (LC)). We compared the immune response of LC and individuals with post-COVID full recovery (HC) during the Omicron pandemic. Two hundred ninety-two patients with confirmed COVID infections from January to May 2022 were enrolled. We observed anti-SARS-CoV-2 receptor-binding domain immunoglobulin G, surrogate virus neutralization test, T cell subsets, and neutralizing antibodies against Wuhan, BA.1, and BA.5 viruses (NeuT). NeuT was markedly reduced against BA.1 and BA.5 in HC and LC groups, while antibodies were more sustained with three doses and an updated booster shot than ≤2-dose vaccinations. The viral neutralization ability declined at >84-days after COVID-19 onset (PC) in both groups. PD1-expressed central and effector memory CD4+ T cells, and central memory CD8+ T cells were reduced in the first months PC in LC. Therefore, booster vaccines may be required sooner after the most recent infection to rescue T cell function for people with symptomatic LC.
Collapse
Affiliation(s)
- Tanyaporn Pongkunakorn
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Thamonwan Manosan
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Apinya Surawit
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Suphawan Ophakas
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Pichanun Mongkolsucharitkul
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Sureeporn Pumeiam
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Sophida Suta
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Bonggochpass Pinsawas
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| | - Nitat Sookrung
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (N.S.); (N.S.); (K.M.); (P.P.); (A.T.)
| | - Nawannaporn Saelim
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (N.S.); (N.S.); (K.M.); (P.P.); (A.T.)
| | - Kodchakorn Mahasongkram
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (N.S.); (N.S.); (K.M.); (P.P.); (A.T.)
| | - Pannathee Prangtaworn
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (N.S.); (N.S.); (K.M.); (P.P.); (A.T.)
| | - Anchalee Tungtrongchitr
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (N.S.); (N.S.); (K.M.); (P.P.); (A.T.)
| | - Watip Tangjittipokin
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (W.T.); (S.M.); (K.B.)
| | - Suthee Mangmee
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (W.T.); (S.M.); (K.B.)
| | - Kobporn Boonnak
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (W.T.); (S.M.); (K.B.)
| | - Tassanee Narkdontri
- Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.N.); (N.T.)
| | - Nipaporn Teerawattanapong
- Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.N.); (N.T.)
| | - Rungsima Wanitphadeedecha
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Korapat Mayurasakorn
- Siriraj Population Health and Nutrition Research Group, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.P.); (T.M.); (A.S.); (S.O.); (P.M.); (S.P.); (S.S.); (B.P.)
| |
Collapse
|
29
|
Joo V, Petrovas C, de Leval L, Noto A, Obeid M, Fenwick C, Pantaleo G. A CD64/FcγRI-mediated mechanism hijacks PD-1 from PD-L1/2 interaction and enhances anti-PD-1 functional recovery of exhausted T cells. Front Immunol 2023; 14:1213375. [PMID: 37622123 PMCID: PMC10446174 DOI: 10.3389/fimmu.2023.1213375] [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: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAb) targeting the immune checkpoint inhibitor programmed cell death protein 1 (PD-1) have achieved considerable clinical success in anti-cancer therapy through relieving T cell exhaustion. Blockade of PD-1 interaction with its ligands PD-L1 and PD-L2 is an important determinant in promoting the functional recovery of exhausted T cells. Here, we show that anti-PD-1 mAbs act through an alternative mechanism leading to the downregulation of PD-1 surface expression on memory CD4+ and CD8+ T cells. PD-1 receptor downregulation is a distinct process from receptor endocytosis and occurs in a CD14+ monocyte dependent manner with the CD64/Fcγ receptor I acting as the primary factor for this T cell extrinsic process. Importantly, downregulation of surface PD-1 strongly enhances antigen-specific functional recovery of exhausted PD-1+CD8+ T cells. Our study demonstrates a novel mechanism for reducing cell surface levels of PD-1 and limiting the inhibitory targeting by PD-L1/2 and thereby enhancing the efficacy of anti-PD-1 Ab in restoring T cell functionality.
Collapse
Affiliation(s)
- Victor Joo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Constantinos Petrovas
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Michel Obeid
- Lausanne Center for Immuno-oncology Toxicities (LCIT), Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
Ma H, Ren S, Meng Q, Su B, Wang K, Liu Y, Wang J, Ding D, Li X. Role of Tim-3 in COVID-19: a potential biomarker and therapeutic target. Arch Virol 2023; 168:213. [PMID: 37522944 DOI: 10.1007/s00705-023-05842-2] [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: 01/18/2023] [Accepted: 06/16/2023] [Indexed: 08/01/2023]
Abstract
T cell immunoglobulin and mucin domain containing protein 3 (Tim-3), an immune checkpoint, is important for maintaining immune tolerance. There is increasing evidence that Tim-3 is aberrantly expressed in patients with COVID-19, indicating that it may play an important role in COVID-19. In this review, we discuss the altered expression and potential role of Tim-3 in COVID-19. The expression of Tim-3 and its soluble form (sTim-3) has been found to be upregulated in COVID-19 patients. The levels of Tim-3 on T cells and circulating sTim-3 have been shown to be associated with the severity of COVID-19, suggesting that this protein could be a potential biomarker of COVID-19. Moreover, this review also highlights the potential of Tim-3 as a therapeutic target of COVID-19.
Collapse
Affiliation(s)
- Haodong Ma
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Shengju Ren
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Qingpeng Meng
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Boyuan Su
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Kun Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - YiChen Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Junpeng Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, 450003, Zhengzhou, Henan, China.
| | - Degang Ding
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, 450003, Zhengzhou, Henan, China.
| | - Xin Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China.
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, 450001, Zhengzhou, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
31
|
Damiano RF, Rocca CCDA, Serafim ADP, Loftis JM, Talib LL, Pan PM, Cunha-Neto E, Kalil J, de Castro GS, Seelaender M, Guedes BF, Nagahashi Marie SK, de Souza HP, Nitrini R, Miguel EC, Busatto G, Forlenza OV. Cognitive impairment in long-COVID and its association with persistent dysregulation in inflammatory markers. Front Immunol 2023; 14:1174020. [PMID: 37287969 PMCID: PMC10242059 DOI: 10.3389/fimmu.2023.1174020] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/10/2023] [Indexed: 06/09/2023] Open
Abstract
Objective To analyze the potential impact of sociodemographic, clinical and biological factors on the long-term cognitive outcome of patients who survived moderate and severe forms of COVID-19. Methods We assessed 710 adult participants (Mean age = 55 ± 14; 48.3% were female) 6 to 11 months after hospital discharge with a complete cognitive battery, as well as a psychiatric, clinical and laboratory evaluation. A large set of inferential statistical methods was used to predict potential variables associated with any long-term cognitive impairment, with a focus on a panel of 28 cytokines and other blood inflammatory and disease severity markers. Results Concerning the subjective assessment of cognitive performance, 36.1% reported a slightly poorer overall cognitive performance, and 14.6% reported being severely impacted, compared to their pre-COVID-19 status. Multivariate analysis found sex, age, ethnicity, education, comorbidity, frailty and physical activity associated with general cognition. A bivariate analysis found that G-CSF, IFN-alfa2, IL13, IL15, IL1.RA, EL1.alfa, IL45, IL5, IL6, IL7, TNF-Beta, VEGF, Follow-up C-Reactive Protein, and Follow-up D-Dimer were significantly (p<.05) associated with general cognition. However, a LASSO regression that included all follow-up variables, inflammatory markers and cytokines did not support these findings. Conclusion Though we identified several sociodemographic characteristics that might protect against cognitive impairment following SARS-CoV-2 infection, our data do not support a prominent role for clinical status (both during acute and long-stage of COVID-19) or inflammatory background (also during acute and long-stage of COVID-19) to explain the cognitive deficits that can follow COVID-19 infection.
Collapse
Affiliation(s)
- Rodolfo Furlan Damiano
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Cristiana Castanho de Almeida Rocca
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | | | - Jennifer M. Loftis
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States
- Departments of Psychiatry and Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Leda Leme Talib
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Pedro Mário Pan
- Departamento de Psiquiatria, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Edecio Cunha-Neto
- Departamento de Cínica Médica, Universidade de São Paulo FMUSP, São Paulo, SP, Brazil
- Institute for Investigation in Immunology/National Institutes for Science and Technology (iii/INCT), São Paulo, Brazil
| | - Jorge Kalil
- Departamento de Cínica Médica, Universidade de São Paulo FMUSP, São Paulo, SP, Brazil
- Institute for Investigation in Immunology/National Institutes for Science and Technology (iii/INCT), São Paulo, Brazil
| | - Gabriela Salim de Castro
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, SP, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, SP, Brazil
| | - Bruno F. Guedes
- Departamento de Neurologia, Universidade de São Paulo FMUSP, São Paulo, Brazil
| | | | | | - Ricardo Nitrini
- Departamento de Neurologia, Universidade de São Paulo FMUSP, São Paulo, Brazil
| | - Euripedes Constantino Miguel
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Geraldo Busatto
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Orestes V. Forlenza
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| |
Collapse
|
32
|
Petersen E, Chudakova D, Erdyneeva D, Zorigt D, Shabalina E, Gudkov D, Karalkin P, Reshetov I, Mynbaev OA. COVID-19-The Shift of Homeostasis into Oncopathology or Chronic Fibrosis in Terms of Female Reproductive System Involvement. Int J Mol Sci 2023; 24:ijms24108579. [PMID: 37239926 DOI: 10.3390/ijms24108579] [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: 01/17/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 coronavirus remains a global public health concern due to the systemic nature of the infection and its long-term consequences, many of which remain to be elucidated. SARS-CoV-2 targets endothelial cells and blood vessels, altering the tissue microenvironment, its secretion, immune-cell subpopulations, the extracellular matrix, and the molecular composition and mechanical properties. The female reproductive system has high regenerative potential, but can accumulate damage, including due to SARS-CoV-2. COVID-19 is profibrotic and can change the tissue microenvironment toward an oncogenic niche. This makes COVID-19 and its consequences one of the potential regulators of a homeostasis shift toward oncopathology and fibrosis in the tissues of the female reproductive system. We are looking at SARS-CoV-2-induced changes at all levels in the female reproductive system.
Collapse
Affiliation(s)
- Elena Petersen
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Daria Chudakova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Daiana Erdyneeva
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Dulamsuren Zorigt
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | | | - Denis Gudkov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Pavel Karalkin
- P.A. Herzen Moscow Research Institute of Oncology, 125284 Moscow, Russia
- Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Igor Reshetov
- Institute of Cluster Oncology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ospan A Mynbaev
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| |
Collapse
|
33
|
Montefusco L, Pastore I, Lunati ME, Fiorina P. Increased Risk of Diabetes and Diabetic Ketoacidosis Associated With COVID-19. Diabetes 2023; 72:560-561. [PMID: 37146280 DOI: 10.2337/db22-0908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/20/2023] [Indexed: 05/07/2023]
Affiliation(s)
- Laura Montefusco
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | | | - Paolo Fiorina
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
34
|
Kenny G, Townsend L, Savinelli S, Mallon PWG. Long COVID: Clinical characteristics, proposed pathogenesis and potential therapeutic targets. Front Mol Biosci 2023; 10:1157651. [PMID: 37179568 PMCID: PMC10171433 DOI: 10.3389/fmolb.2023.1157651] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023] Open
Abstract
The emergence of persistent ill-health in the aftermath of SARS-CoV-2 infection has presented significant challenges to patients, healthcare workers and researchers. Termed long COVID, or post-acute sequelae of COVID-19 (PASC), the symptoms of this condition are highly variable and span multiple body systems. The underlying pathophysiology remains poorly understood, with no therapeutic agents proven to be effective. This narrative review describes predominant clinical features and phenotypes of long COVID alongside the data supporting potential pathogenesis of these phenotypes including ongoing immune dysregulation, viral persistence, endotheliopathy, gastrointestinal microbiome disturbance, autoimmunity, and dysautonomia. Finally, we describe current potential therapies under investigation, as well as future potential therapeutic options based on the proposed pathogenesis research.
Collapse
Affiliation(s)
- Grace Kenny
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| | - Liam Townsend
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| | - Stefano Savinelli
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Dublin, Ireland
| |
Collapse
|
35
|
Pastukhova E, Ghazawi FM. Eruptive halo nevi and new-onset vitiligo post-COVID-19 infection. JAAD Case Rep 2023; 34:43-44. [PMID: 36816762 PMCID: PMC9927811 DOI: 10.1016/j.jdcr.2023.01.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 02/16/2023] Open
Affiliation(s)
- Elena Pastukhova
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Feras M Ghazawi
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Division of Dermatology, University of Ottawa, and The Ottawa Hospital, Ottawa, Ontario, Canada
| |
Collapse
|
36
|
Tanvetyanon T, Chen DT, Gray JE. Impact of COVID-19 Pandemic on Frontline Pembrolizumab-Based Treatment for Advanced Lung Cancer. J Clin Med 2023; 12:jcm12041611. [PMID: 36836146 PMCID: PMC9960275 DOI: 10.3390/jcm12041611] [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: 12/20/2022] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Pembrolizumab monotherapy or pembrolizumab plus chemotherapy has become an important frontline treatment for advanced non-small cell lung cancer (NSCLC). To date, it remains unclear how the coronavirus disease 2019 (COVID-19) pandemic impacted the treatment outcome. METHODS A quasi-experimental study was conducted based on a real-world database, comparing pandemic with pre-pandemic patient cohorts. The pandemic cohort consisted of patients who initiated treatment from March to July 2020, with follow-up through March 2021. The pre-pandemic cohort consisted of those initiating treatment between March and July 2019.The outcome was overall real-world survival. Multivariable Cox-proportional hazard models were constructed. RESULTS Analyses included data from 2090 patients: 998 in the pandemic cohort and 1092 in the pre-pandemic cohort. Baseline characteristics were comparable, with 33% of patients having PD-L1 expression level ≥50% and 29% of patients receiving pembrolizumab monotherapy. Among those treated with pembrolizumab monotherapy (N = 613), there was a differential impact of the pandemic on survival by PD-L1 expression levels (p-interaction = 0.02). For those with PD-L1 level < 50%, survival was better in the pandemic cohort than the pre-pandemic cohort: hazard ratio (HR) 0.64 (95% CI: 0.43-0.97, p = 0.03). However, for those with PD-L1 level ≥ 50%, survival was not better in the pandemic cohort: HR 1.17 (95% CI: 0.85-1.61, p = 0.34). We found no statistically significant impact of the pandemic on survival among patients treated with pembrolizumab plus chemotherapy. CONCLUSIONS The COVID-19 pandemic was associated with an increase in survival among patients with lower PD-L1 expression who were treated with pembrolizumab monotherapy. This finding suggests an increased efficacy of immunotherapy due to viral exposure in this population.
Collapse
|
37
|
Employing T-Cell Memory to Effectively Target SARS-CoV-2. Pathogens 2023; 12:pathogens12020301. [PMID: 36839573 PMCID: PMC9967959 DOI: 10.3390/pathogens12020301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
Well-trained T-cell immunity is needed for early viral containment, especially with the help of an ideal vaccine. Although most severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected convalescent cases have recovered with the generation of virus-specific memory T cells, some cases have encountered T-cell abnormalities. The emergence of several mutant strains has even threatened the effectiveness of the T-cell immunity that was established with the first-generation vaccines. Currently, the development of next-generation vaccines involves trying several approaches to educate T-cell memory to trigger a broad and fast response that targets several viral proteins. As the shaping of T-cell immunity in its fast and efficient form becomes important, this review discusses several interesting vaccine approaches to effectively employ T-cell memory for efficient viral containment. In addition, some essential facts and future possible consequences of using current vaccines are also highlighted.
Collapse
|
38
|
Roe K. Accelerated T-cell exhaustion: its potential role in autoimmune disease and hyperinflammatory disease pathogenesis. Hum Cell 2023; 36:866-869. [PMID: 36478089 PMCID: PMC9735067 DOI: 10.1007/s13577-022-00839-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
|
39
|
Huang SF, Ying-Jung Wu A, Shin-Jung Lee S, Huang YS, Lee CY, Yang TL, Wang HW, Chen HJ, Chen YC, Ho TS, Kuo CF, Lin YT. COVID-19 associated mold infections: Review of COVID-19 associated pulmonary aspergillosis and mucormycosis. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022:S1684-1182(22)00285-7. [PMID: 36586744 PMCID: PMC9751001 DOI: 10.1016/j.jmii.2022.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
COVID-19-associated mold infection (CAMI) is defined as development of mold infections in COVID-19 patients. Co-pathogenesis of viral and fungal infections include the disruption of tissue barrier following SARS CoV-2 infection with the damage in the alveolar space, respiratory epithelium and endothelium injury and overwhelming inflammation and immune dysregulation during severe COVID-19. Other predisposing risk factors permissive to fungal infections during COVID-19 include the administration of immune modulators such as corticosteroids and IL-6 antagonist. COVID-19-associated pulmonary aspergillosis (CAPA) and COVID-19-associated mucormycosis (CAM) is increasingly reported during the COVID-19 pandemic. CAPA usually developed within the first month of COVID infection, and CAM frequently arose 10-15 days post diagnosis of COVID-19. Diagnosis is challenging and often indistinguishable during the cytokine storm in COVID-19, and several diagnostic criteria have been proposed. Development of CAPA and CAM is associated with a high mortality despiteappropriate anti-mold therapy. Both isavuconazole and amphotericin B can be used for treatment of CAPA and CAM; voriconazole is the primary agent for CAPA and posaconazole is an alternative for CAM. Aggressive surgery is recommended for CAM to improve patient survival. A high index of suspicion and timely and appropriate treatment is crucial to improve patient outcome.
Collapse
Affiliation(s)
- Shiang-Fen Huang
- Division of Infectious Disease, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan,School of Internal Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan
| | - Alice Ying-Jung Wu
- Division of Infectious Diseases, Department of Medicine, MacKay Memorial Hospital, Taipei, Taiwan,MacKay Medical College, New Taipei City, Taiwan
| | - Susan Shin-Jung Lee
- School of Internal Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan,Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Taiwan
| | - Yu-Shan Huang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Yuan Lee
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Te-Liang Yang
- Department of Pediatrics, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan,Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Hsiao-Wei Wang
- Division of Infectious Diseases, Department of Internal Medicine, Shin Kong Wu Ho- Su Memorial Hospital, Taipei, Taiwan
| | - Hung Jui Chen
- Department of Infectious Diseases, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yi Ching Chen
- Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan,College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzong-Shiann Ho
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan,Department of Pediatrics, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| | - Chien-Feng Kuo
- Division of Infectious Diseases, Department of Medicine, MacKay Memorial Hospital, Taipei, Taiwan,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan,Corresponding author
| | - Yi-Tsung Lin
- Division of Infectious Disease, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan,Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan,Corresponding author
| | | |
Collapse
|
40
|
Alahdal M, Elkord E. Exhaustion and over-activation of immune cells in COVID-19: Challenges and therapeutic opportunities. Clin Immunol 2022; 245:109177. [PMID: 36356848 PMCID: PMC9640209 DOI: 10.1016/j.clim.2022.109177] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
Exhaustion of immune cells in COVID-19 remains a serious concern for infection management and therapeutic interventions. As reported, immune cells such as T effector cells (Teff), T regulatory cells (Tregs), natural killer cells (NKs), and antigen-presenting cells (APCs) exhibit uncontrolled functions in COVID-19. Unfortunately, the mechanisms that orchestrate immune cell functionality and virus interaction are still unknown. Recent studies linked adaptive immune cell exhaustion to underlying epigenetic mechanisms that regulate the epigenetic transcription of inhibitory immune checkpoint receptors (ICs). Further to that, the over-activation of T cells accompanied by the dysfunctionality of DCs and Tregs may enhance uncontrollable alveoli inflammation and cytokine storm in COVID-19. This might explain the reasons behind the failure of DC-based vaccines in inducing sufficient anti-viral responses. This review explains the processes behind the over-activation and exhaustion of innate and adaptive immune cells in COVID-19, which may contribute to developing novel immune intervention strategies.
Collapse
Affiliation(s)
- Murad Alahdal
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33 Birkat Al Mouz, Nizwa 616, Oman.
| | - Eyad Elkord
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33 Birkat Al Mouz, Nizwa 616, Oman; Department of Biological Sciences and Chemistry, Faculty of Arts and Sciences, University of Nizwa, Birkat Al Mouz, Nizwa 616, Oman; Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom.
| |
Collapse
|
41
|
Trøseid M, Dahl TB, Holter JC, Kildal AB, Murphy SL, Yang K, Quiles-Jiménez A, Heggelund L, Müller KE, Tveita A, Michelsen AE, Bøe S, Holten AR, Hoel H, Mathiessen A, Aaløkken TM, Fevang B, Granerud BK, Tonby K, Henriksen KN, Lerum TV, Müller F, Skjønsberg OH, Barratt-Due A, Dyrhol-Riise AM, Aukrust P, Halvorsen B, Ueland T. Persistent T-cell exhaustion in relation to prolonged pulmonary pathology and death after severe COVID-19: Results from two Norwegian cohort studies. J Intern Med 2022; 292:816-828. [PMID: 35982589 PMCID: PMC9805032 DOI: 10.1111/joim.13549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND T-cell activation is associated with an adverse outcome in COVID-19, but whether T-cell activation and exhaustion relate to persistent respiratory dysfunction and death is unknown. OBJECTIVES To investigate whether T-cell activation and exhaustion persist and are associated with prolonged respiratory dysfunction and death after hospitalization for COVID-19. METHODS Plasma and serum from two Norwegian cohorts of hospitalized patients with COVID-19 (n = 414) were analyzed for soluble (s) markers of T-cell activation (sCD25) and exhaustion (sTim-3) during hospitalization and follow-up. RESULTS Both markers were strongly associated with acute respiratory failure, but only sTim-3 was independently associated with 60-day mortality. Levels of sTim-3 remained elevated 3 and 12 months after hospitalization and were associated with pulmonary radiological pathology after 3 months. CONCLUSION Our findings suggest prolonged T-cell exhaustion is an important immunological sequela, potentially related to long-term outcomes after severe COVID-19.
Collapse
Affiliation(s)
- Marius Trøseid
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tuva B Dahl
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Division of Critical Care and Emergencies, Oslo University Hospital, Oslo, Norway
| | - Jan C Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Anders B Kildal
- Department of Anesthesiology and Intensive Care, University Hospital of North Norway, Tromsø, Norway
| | - Sarah L Murphy
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kuan Yang
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ana Quiles-Jiménez
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Heggelund
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway.,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Karl Erik Müller
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Anders Tveita
- Department of Internal Medicine, Baerum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway.,Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Annika E Michelsen
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Simen Bøe
- Department of Anesthesiology and Intensive Care, Hammerfest County Hospital, Hammerfest, Norway
| | - Aleksander R Holten
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Acute Medicine, Oslo University Hospital, Oslo, Norway
| | - Hedda Hoel
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Internal Medicine, Lovisenberg Diakonal Hospital, Oslo, Norway
| | | | - Trond M Aaløkken
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Radiology and Nuclear Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Børre Fevang
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Beathe K Granerud
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Katerina N Henriksen
- Hospital Pharmacies, South-Eastern Norway Enterprise, Oslo, Norway.,Department of Hematology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tøri V Lerum
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pulmonary Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Fredrik Müller
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Ole H Skjønsberg
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pulmonary Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Andreas Barratt-Due
- Division of Critical Care and Emergencies, Oslo University Hospital, Oslo, Norway.,Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Anne M Dyrhol-Riise
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Pål Aukrust
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Clinical Medicine, Thrombosis Research and Expertise Center (TREC), UiT-The Arctic University of Norway, Tromsø, Norway
| | | | | |
Collapse
|
42
|
Monroy-Iglesias MJ, Tremble K, Russell B, Moss C, Dolly S, Sita-Lumsden A, Cortellini A, Pinato DJ, Rigg A, Karagiannis SN, Van Hemelrijck M. Long-term effects of COVID-19 on cancer patients: the experience from Guy's Cancer Centre. Future Oncol 2022; 18:3585-3594. [PMID: 36172860 DOI: 10.2217/fon-2022-0088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Few studies have investigated the long-term effects of COVID-19 on cancer patients. Materials & methods: The authors conducted a telephone survey on the long-term symptoms of cancer patients from Guy's Cancer Centre. They compared patients whose symptoms occurred/got worse over 4 weeks after COVID-19 diagnosis (classified as long COVID) with patients who did not develop symptoms or whose symptoms occurred/got worse in the first 4 weeks after diagnosis. Results: The authors analyzed responses from 80 patients with a previous COVID-19 diagnosis; 51.3% (n = 41) developed long COVID. The most common symptoms were fatigue, breathlessness and cognitive impairment. Conclusion: Findings suggest that over half of the cancer population will experience long-term effects after their initial COVID-19 diagnosis. Further studies are required to validate the findings of this study.
Collapse
Affiliation(s)
- Maria J Monroy-Iglesias
- King's College London, School of Cancer & Pharmaceutical Sciences, Translational Oncology & Urology Research (TOUR), London, SE1 9RT, UK
| | - Kathryn Tremble
- Medical Oncology, Guy's & St. Thomas' NHS Foundation Trust (GSTT), London, SE1 9RT, UK
| | - Beth Russell
- King's College London, School of Cancer & Pharmaceutical Sciences, Translational Oncology & Urology Research (TOUR), London, SE1 9RT, UK
| | - Charlotte Moss
- King's College London, School of Cancer & Pharmaceutical Sciences, Translational Oncology & Urology Research (TOUR), London, SE1 9RT, UK
| | - Saoirse Dolly
- Medical Oncology, Guy's & St. Thomas' NHS Foundation Trust (GSTT), London, SE1 9RT, UK
| | - Ailsa Sita-Lumsden
- Medical Oncology, Guy's & St. Thomas' NHS Foundation Trust (GSTT), London, SE1 9RT, UK
| | - Alessio Cortellini
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London, W120HS, UK
| | - David James Pinato
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London, W120HS, UK
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Anne Rigg
- Medical Oncology, Guy's & St. Thomas' NHS Foundation Trust (GSTT), London, SE1 9RT, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, SE1 9RT, UK
| | - Mieke Van Hemelrijck
- King's College London, School of Cancer & Pharmaceutical Sciences, Translational Oncology & Urology Research (TOUR), London, SE1 9RT, UK
| |
Collapse
|
43
|
Ravaglia C, Doglioni C, Chilosi M, Piciucchi S, Dubini A, Rossi G, Pedica F, Puglisi S, Donati L, Tomassetti S, Poletti V. Clinical, radiological and pathological findings in patients with persistent lung disease following SARS-CoV-2 infection. Eur Respir J 2022; 60:2102411. [PMID: 35301248 PMCID: PMC8932282 DOI: 10.1183/13993003.02411-2021] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 02/13/2022] [Indexed: 12/18/2022]
Abstract
Some patients experience pulmonary sequelae after SARS-CoV-2 infection, ranging from self-limited abnormalities to major lung diseases. Morphological analysis of lung tissue may help our understanding of pathogenic mechanisms and help to provide consistent personalised management. The aim of this study was to ascertain morphological and immunomolecular features of lung tissue. Transbronchial lung cryobiopsy was carried out in patients with persistent symptoms and computed tomography suggestive of residual lung disease after recovery from SARS-CoV-2 infection. 164 patients were referred for suspected pulmonary sequelae after COVID-19; 10 patients with >5% parenchymal lung disease underwent lung biopsy. The histological pattern of lung disease was not homogeneous and three different case clusters could be identified, which was mirrored by their clinical and radiological features. Cluster 1 ("chronic fibrosing") was characterised by post-infection progression of pre-existing interstitial pneumonias. Cluster 2 ("acute/subacute injury") was characterised by different types and grades of lung injury, ranging from organising pneumonia and fibrosing nonspecific interstitial pneumonia to diffuse alveolar damage. Cluster 3 ("vascular changes") was characterised by diffuse vascular increase, dilatation and distortion (capillaries and venules) within otherwise normal parenchyma. Clusters 2 and 3 had immunophenotypical changes similar to those observed in early/mild COVID-19 pneumonias (abnormal expression of STAT3 in hyperplastic pneumocytes and PD-L1, IDO and STAT3 in endothelial cells). This is the first study correlating histological/immunohistochemical patterns with clinical and radiological pictures of patients with post-COVID lung disease. Different phenotypes with potentially different underlying pathogenic mechanisms have been identified.
Collapse
Affiliation(s)
- Claudia Ravaglia
- Dept of Thoracic Diseases, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Claudio Doglioni
- Dept of Pathology, University Vita-Salute, Milan and San Raffaele Scientific Institute, Milan, Italy
| | - Marco Chilosi
- Dept of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Sara Piciucchi
- Dept of Radiology, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Alessandra Dubini
- Dept of Pathology, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Giulio Rossi
- Dept of Pathology, Fondazione Poliambulanza Istituto Ospedaliero Multispecialistico, Brescia, Italy
| | - Federica Pedica
- Dept of Pathology, San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Puglisi
- Dept of Thoracic Diseases, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
| | - Luca Donati
- Biostatistics and Clinical Trial Unit, Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori"-IRST S.r.l., IRCCS, Meldola, Italy
| | - Sara Tomassetti
- Dept of Experimental and Clinical Medicine, Careggi University Hospital, Firenze, Italy
| | - Venerino Poletti
- Dept of Thoracic Diseases, G.B. Morgagni Hospital/University of Bologna, Forlì, Italy
- DIMES, University of Bologna, Bologna, Italy
- Dept of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
44
|
Bolla AM, Loretelli C, Montefusco L, Finzi G, Abdi R, Ben Nasr M, Lunati ME, Pastore I, Bonventre JV, Nebuloni M, Rusconi S, Santus P, Zuccotti G, Galli M, D’Addio F, Fiorina P. Inflammation and vascular dysfunction: The negative synergistic combination of diabetes and COVID-19. Diabetes Metab Res Rev 2022; 38:e3565. [PMID: 35830597 PMCID: PMC9349661 DOI: 10.1002/dmrr.3565] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/08/2023]
Abstract
AIMS Several reports indicate that diabetes determines an increased mortality risk in patients with coronavirus disease 19 (COVID-19) and a good glycaemic control appears to be associated with more favourable outcomes. Evidence also supports that COVID-19 pneumonia only accounts for a part of COVID-19 related deaths. This disease is indeed characterised by abnormal inflammatory response and vascular dysfunction, leading to the involvement and failure of different systems, including severe acute respiratory distress syndrome, coagulopathy, myocardial damage and renal failure. Inflammation and vascular dysfunction are also well-known features of hyperglycemia and diabetes, making up the ground for a detrimental synergistic combination that could explain the increased mortality observed in hyperglycaemic patients. MATERIALS AND METHODS In this work, we conduct a narrative review on this intriguing connection. Together with this, we also present the clinical characteristics, outcomes, laboratory and histopathological findings related to this topic of a cohort of nearly 1000 subjects with COVID-19 admitted to a third-level Hospital in Milan. RESULTS We found an increased mortality in subjects with COVID-19 and diabetes, together with an altered inflammatory profile. CONCLUSIONS This may support the hypothesis that diabetes and COVID-19 meet at the crossroads of inflammation and vascular dysfunction. (ClinicalTrials.gov NCT04463849 and NCT04382794).
Collapse
Affiliation(s)
| | - Cristian Loretelli
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
| | | | | | - Reza Abdi
- Nephrology DivisionBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Moufida Ben Nasr
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Ida Pastore
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
| | - Joseph V. Bonventre
- Nephrology DivisionBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Manuela Nebuloni
- Pathology UnitASST Fatebenefratelli‐SaccoMilanItaly
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
| | - Stefano Rusconi
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
| | - Pierachille Santus
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
- Division of Respiratory DiseasesASST Fatebenefratelli‐SaccoMilanItaly
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
- Department of Pediatrics“V. Buzzi” Children's HospitalMilanItaly
| | - Massimo Galli
- Department of Biomedical and Clinical Sciences L. SaccoUniversity of MilanMilanItaly
- III Division of Infectious DiseasesLuigi Sacco HospitalASST Fatebenefratelli‐SaccoMilanItaly
| | - Francesca D’Addio
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
| | - Paolo Fiorina
- Division of EndocrinologyASST Fatebenefratelli‐SaccoMilanItaly
- International Center for T1DPediatric Clinical Research Center Romeo ed Enrica InvernizziDepartment of Biomedical and Clinical Science L. SaccoUniversity of MilanMilanItaly
- Nephrology DivisionBoston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
45
|
Rienzo M, Skirecki T, Monneret G, Timsit JF. Immune checkpoint inhibitors for the treatment of sepsis:insights from preclinical and clinical development. Expert Opin Investig Drugs 2022; 31:885-894. [PMID: 35944174 DOI: 10.1080/13543784.2022.2102477] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Sepsis represents one-fifth of all deaths worldwide and is associated with huge costs. Regarding disease progression, it is now well established that sepsis induces a state of acquired immunosuppression, with an increased risk of secondary infections that contributes to patients' worsening. Thus, tackling sepsis-induced immunosuppression represents a promising perspective. AREAS COVERED Of mechanisms responsible for sepsis-induced immunosuppression, the increased expression of co-inhibitory receptors (aka immune checkpoint) such as PD-1, CTLA4, TIM-3, LAG-3 or BTLA and their ligands recently received considerable interest since their inhibition, thanks to the so-called checkpoint inhibitors (CPI), provided astonishing results in cancer by rebooting immune functions. This review reports on the first landmarks of these molecules in sepsis. We introduce them in terms of basic immunology in line with sepsis pathophysiology both in experimental models and observational works and assess the first human clinical studies. EXPERT OPINION Preclinical results are positive and the first human clinical trials, although currently limited to the early phase, showed a beneficial effect on immunological functions and/or markers and suggested that tolerance of CPIs side effects, mainly auto-immune disorders, is acceptable in sepsis. Elsewhere, in some specific infections leading to ICU admission (or occurring during ICU stay), such as fungal infections, preliminary convincing case reports have been published. Overall, the first results regarding CPIs in sepsis appear encouraging. However, further efforts are warranted, especially in defining the right patients to be treated (i.e., in an individualized approach) and establishing the optimal time to start an immune restoration. Larger trials are now mandatory to confirm CPIs' potential in sepsis.
Collapse
Affiliation(s)
- Mario Rienzo
- AP-HP, Bichat Hospital, Medical and infectious diseases ICU (MI2), F-75018 Paris, France
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Guillaume Monneret
- Immunology Laboratory, Hôpital E. Herriot, Hospices Civils de Lyon, Lyon, F-69003.,Université de Lyon, EA7426, Hôpital E. Herriot, Lyon, F-69003
| | - Jean-François Timsit
- AP-HP, Bichat Hospital, Medical and infectious diseases ICU (MI2), F-75018 Paris, France.,University of Paris, IAME, INSERM, F-75018 Paris, France
| |
Collapse
|
46
|
D'Addio F, Sabiu G, Usuelli V, Assi E, Abdelsalam A, Maestroni A, Seelam AJ, Ben Nasr M, Loretelli C, Mileto D, Rossi G, Pastore I, Montefusco L, Morpurgo PS, Plebani L, Rossi A, Chebat E, Bolla AM, Lunati ME, Mameli C, Macedoni M, Antinori S, Rusconi S, Gallieni M, Berra C, Folli F, Galli M, Gismondo MR, Zuccotti G, Fiorina P. Immunogenicity and Safety of SARS-CoV-2 mRNA Vaccines in a Cohort of Patients With Type 1 Diabetes. Diabetes 2022; 71:1800-1806. [PMID: 35551366 DOI: 10.2337/db22-0053] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/30/2022] [Indexed: 11/13/2022]
Abstract
Patients with type 1 diabetes (T1D) may develop severe outcomes during coronavirus disease 2019 (COVID-19), but their ability to generate an immune response against the SARS-CoV-2 mRNA vaccines remains to be established. We evaluated the safety, immunogenicity, and glycometabolic effects of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines in patients with T1D. A total of 375 patients (326 with T1D and 49 subjects without diabetes) who received two doses of the SARS-CoV-2 mRNA vaccines (mRNA-1273, BNT162b2) between March and April 2021 at ASST Fatebenefratelli Sacco were included in this monocentric observational study. Local and systemic adverse events were reported in both groups after SARS-CoV-2 mRNA vaccination, without statistical differences between them. While both patients with T1D and subjects without diabetes exhibited a parallel increase in anti-SARS-CoV-2 spike titers after vaccination, the majority of patients with T1D (70% and 78%, respectively) did not show any increase in the SARS-CoV-2-specific cytotoxic response compared with the robust increase observed in all subjects without diabetes. A reduced secretion of the T-cell-related cytokines interleukin-2 and tumor necrosis factor-α in vaccinated patients with T1D was also observed. No glycometabolic alterations were evident in patients with T1D using continuous glucose monitoring during follow-up. Administration of the SARS-CoV-2 mRNA vaccine is associated with an impaired cellular SARS-CoV-2-specific cytotoxic immune response in patients with T1D.
Collapse
Affiliation(s)
- Francesca D'Addio
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Gianmarco Sabiu
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
- Nephrology and Dialysis Unit, ASST Fatebenefratelli Sacco, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Vera Usuelli
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Emma Assi
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Ahmed Abdelsalam
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Anna Maestroni
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Andy Joe Seelam
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Moufida Ben Nasr
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Cristian Loretelli
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Davide Mileto
- Diagnostic Services, Clinical Microbiology, Virology and Bioemergence Diagnostics, ASST Fatebenefratelli Sacco, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Giada Rossi
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Ida Pastore
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Laura Montefusco
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Paola S Morpurgo
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Laura Plebani
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Antonio Rossi
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Enrica Chebat
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Andrea M Bolla
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
| | | | - Chiara Mameli
- Pediatric Department, Buzzi Children's Hospital, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Maddalena Macedoni
- Pediatric Department, Buzzi Children's Hospital, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Spinello Antinori
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Stefano Rusconi
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Maurizio Gallieni
- Nephrology and Dialysis Unit, ASST Fatebenefratelli Sacco, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Cesare Berra
- Metabolic Diseases and Diabetes, Multimedica IRCCS, Sesto San Giovanni, Milan, Italy
| | - Franco Folli
- Endocrinology and Metabolism, Department of Health Science, Università di Milano, ASST Santi Paolo e Carlo, Milan, Italy
| | - Massimo Galli
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Maria Rita Gismondo
- Diagnostic Services, Clinical Microbiology, Virology and Bioemergence Diagnostics, ASST Fatebenefratelli Sacco, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children's Hospital, and Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, Milan, Italy
- Division of Endocrinology, ASST Fatebenefratelli Sacco, Milan, Italy
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
47
|
Goubran H, Stakiw J, Seghatchian J, Ragab G, Burnouf T. SARS-CoV-2 and cancer: the intriguing and informative cross-talk. Transfus Apher Sci 2022; 61:103488. [PMID: 35753906 PMCID: PMC9192107 DOI: 10.1016/j.transci.2022.103488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has significantly disrupted and burdened the diagnostic workup and delivery of care, including transfusion, to cancer patients across the globe. Furthermore, cancer patients suffering from solid tumors or hematologic malignancies were more prone to the infection and had higher morbidity and mortality than the rest of the population. Major signaling pathways have been identified at the intersection of SARS-CoV-2 and cancer cells, often leading to tumor progression or alteration of the tumor response to therapy. The reactivation of oncogenic viruses has also been alluded to in the context and following COVID-19. Paradoxically, certain tumors responded better following the profound infection-induced immune modulation. Unveiling the mechanisms of the virus-tumor cell interactions will lead to a better understanding of the pathophysiology of both cancer progression and virus propagation. It would be challenging to monitor, through the different cancer registries, retrospectively, the response of patients who have been previously exposed to the virus in contrast to those who have not contracted the infection.
Collapse
Affiliation(s)
- Hadi Goubran
- Saskatoon Cancer Centre and College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Julie Stakiw
- Saskatoon Cancer Centre and College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Jerard Seghatchian
- International Consultancy in Blood Components Quality/Safety, Audit/Inspection and DDR Strategy, London, UK
| | - Gaafar Ragab
- Internal Medicine Department, Rheumatology, and Clinical Immunology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt; School of Medicine, Newgiza University (NGU), Giza, Egypt
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
48
|
Abstract
Severe COVID-19 patients display dysregulated expression of checkpoint molecules PD-1 and its ligand PD-L1, suggesting that these checkpoint molecules could be considered as prognostic markers and therapeutic targets in severe cases of COVID-19.
Collapse
Affiliation(s)
- Srinivasa R Bonam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Haitao Hu
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Institute for Human Infections & Immunity, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jagadeesh Bayry
- Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad, 678623, India
| |
Collapse
|
49
|
Wiech M, Chroscicki P, Swatler J, Stepnik D, De Biasi S, Hampel M, Brewinska-Olchowik M, Maliszewska A, Sklinda K, Durlik M, Wierzba W, Cossarizza A, Piwocka K. Remodeling of T Cell Dynamics During Long COVID Is Dependent on Severity of SARS-CoV-2 Infection. Front Immunol 2022; 13:886431. [PMID: 35757700 PMCID: PMC9226563 DOI: 10.3389/fimmu.2022.886431] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022] Open
Abstract
Several COVID-19 convalescents suffer from the post-acute COVID-syndrome (PACS)/long COVID, with symptoms that include fatigue, dyspnea, pulmonary fibrosis, cognitive dysfunctions or even stroke. Given the scale of the worldwide infections, the long-term recovery and the integrative health-care in the nearest future, it is critical to understand the cellular and molecular mechanisms as well as possible predictors of the longitudinal post-COVID-19 responses in convalescent individuals. The immune system and T cell alterations are proposed as drivers of post-acute COVID syndrome. However, despite the number of studies on COVID-19, many of them addressed only the severe convalescents or the short-term responses. Here, we performed longitudinal studies of mild, moderate and severe COVID-19-convalescent patients, at two time points (3 and 6 months from the infection), to assess the dynamics of T cells immune landscape, integrated with patients-reported symptoms. We show that alterations among T cell subsets exhibit different, severity- and time-dependent dynamics, that in severe convalescents result in a polarization towards an exhausted/senescent state of CD4+ and CD8+ T cells and perturbances in CD4+ Tregs. In particular, CD8+ T cells exhibit a high proportion of CD57+ terminal effector cells, together with significant decrease of naïve cell population, augmented granzyme B and IFN-γ production and unresolved inflammation 6 months after infection. Mild convalescents showed increased naïve, and decreased central memory and effector memory CD4+ Treg subsets. Patients from all severity groups can be predisposed to the long COVID symptoms, and fatigue and cognitive dysfunctions are not necessarily related to exhausted/senescent state and T cell dysfunctions, as well as unresolved inflammation that was found only in severe convalescents. In conclusion, the post-COVID-19 functional remodeling of T cells could be seen as a two-step process, leading to distinct convalescent immune states at 6 months after infection. Our data imply that attenuation of the functional polarization together with blocking granzyme B and IFN-γ in CD8+ cells might influence post-COVID alterations in severe convalescents. However, either the search for long COVID predictors or any treatment to prevent PACS and further complications is mandatory in all patients with SARS-CoV-2 infection, and not only in those suffering from severe COVID-19.
Collapse
Affiliation(s)
- Milena Wiech
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Chroscicki
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Dawid Stepnik
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Michal Hampel
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Marta Brewinska-Olchowik
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Maliszewska
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Katarzyna Sklinda
- Department of Radiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marek Durlik
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland.,Departament of Gastroenterological Surgery and Transplantology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Waldemar Wierzba
- Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland.,University of Humanities and Economics, Lodz, Poland
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
50
|
Th1 cytokine endotype discriminates and predicts severe complications in COVID-19. Eur Cytokine Netw 2022; 33:25-36. [PMID: 36266985 PMCID: PMC9595088 DOI: 10.1684/ecn.2022.0477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Treatment of severe and critical cases of coronavirus disease 2019 (COVID-19) is still a top priority in public health. Previously, we reported distinct Th1 cytokines related to the pathophysiology of severe COVID-19 condition. In the present study, we investigated the association of Th1 and Th2 cytokine/chemokine endotypes with cell-mediated immunity via multiplex immunophenotyping, single-cell RNA-Seq analysis of peripheral blood mononuclear cells, and analysis of the clinical features of COVID-19 patients. Based on serum cytokine and systemic inflammatory markers, COVID-19 cases were classified into four clusters of increasing (I-IV) severity. Two prominent clusters were of interest and could be used as prognostic reference for a targeted treatment of severe COVID-19 cases. Cluster III reflected severe/critical pathology and was characterized by decreased in CCL17 levels and increase in IL-6, C-reactive protein CXCL9, IL-18, and IL-10 levels. The second cluster (Cluster II) showed mild to moderate pathology and was characterized by predominated CXCL9 and IL-18 levels, levels of IL-6 and CRP were relatively low. Cluster II patients received anti-inflammatory treatment in early-stage, which may have led prevent disease prognosis which is accompanied to IL-6 and CRP induction. In Cluster III, a decrease in the proportion of effector T cells with signs of T cell exhaustion was observed. This study highlights the mechanisms of endotype clustering based on specific inflammatory markers in related the clinical outcome of COVID-19.
Collapse
|