1
|
Shokrollahi Barough M, Darzi M, Yunesian M, Amini Panah D, Ghane Y, Mottahedan S, Sakinehpour S, Kowsarirad T, Hosseini-Farjam Z, Amirzargar MR, Dehghani S, Shahriyary F, Kabiri MM, Nojomi M, Saraygord-Afshari N, Mostofi SG, Yassin Z, Mojtabavi N. Retrospective analysis of COVID-19 clinical and laboratory data: Constructing a multivariable model across different comorbidities. J Infect Public Health 2024; 17:102566. [PMID: 39454514 DOI: 10.1016/j.jiph.2024.102566] [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: 06/17/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The clinical pathogenesis of COVID-19 necessitates a comprehensive and homogeneous study to understand the disease mechanisms. Identifying clinical symptoms and laboratory parameters as key predictors can guide prognosis and inform effective treatment strategies. This study analyzed comorbidities and laboratory metrics to predict COVID-19 mortality using a homogeneous model. METHOD A retrospective cohort study was conducted on 7500 COVID-19 patients admitted to Rasoul Akram Hospital between 2022 and 2022. Clinical and laboratory data, along with comorbidity information, were collected and analyzed using advanced coding, data alignment, and regression analyses. Machine learning algorithms were employed to identify relevant features and calculate predictive probability scores. RESULTS The frequency and mortality rates of COVID-19 among males (19.3 %) were higher than those among females (17 %) (p = 0.01, OR = 0.85, 95 % CI = 0.76-0.96). Cancer (p < 0.05, OR = 1.9, 95 % CI = 1.48-2.4) and Alzheimer's (p < 0.05, OR = 2.36, 95 % CI = 1.89-2.9) were the two most common comorbidities associated with long-term hospitalization (LTH). Kidney disease (KD) was identified as the most lethal comorbidity (45 % of KD patients) (OR = 5.6, 95 % CI = 5.05-6.04, p < 0.001). Age > 55 was the most predictive parameter for mortality (p < 0.001, OR = 6.5, 95 % CI = 1.03-1.04), and the CT scan score showed no predictive value for death (p > 0.05). WBC, Cr, CRP, ALP, and VBG-HCO3 were the most significant critical data associated with death prediction across all comorbidities (p < 0.05). CONCLUSION COVID-19 is particularly lethal for elderly adults; thus, age plays a crucial role in disease prognosis. Regarding death prediction, various comorbidities rank differently, with KD having a significant impact on mortality outcomes.
Collapse
Affiliation(s)
- Mahdieh Shokrollahi Barough
- Department of Immunology, School of Medicine Iran University of Medical Sciences, Tehran, Iran; Immunology research center institute of immunology and infectious diseases Iran University of Medical Sciences, Tehran, Iran; ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Darzi
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Masoud Yunesian
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Danesh Amini Panah
- Antimicrobial resistance research center, institute of immunology and infectious diseases Iran University of Medical Sciences, Tehran, Iran; Department of Infectious Disease, School of Medicine, Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Yekta Ghane
- Antimicrobial resistance research center, institute of immunology and infectious diseases Iran University of Medical Sciences, Tehran, Iran; Department of Infectious Disease, School of Medicine, Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sam Mottahedan
- Department of Immunology, School of Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Sohrab Sakinehpour
- Radiation Sciences Department, School of paramedicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tahereh Kowsarirad
- Radiation Sciences Department, School of paramedicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Hosseini-Farjam
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Reza Amirzargar
- Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samaneh Dehghani
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Shahriyary
- Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Marzieh Nojomi
- Preventive Medicine and Public Health Research Center, Iran University of Medical Sciences, Iran
| | - Neda Saraygord-Afshari
- Department of Medical Biotechnology, Faculty of Allied Medical sciences Iran University of Medical sciences, Iran
| | - Seyedeh Ghazal Mostofi
- Department of Immunology, School of Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Zeynab Yassin
- Antimicrobial resistance research center, institute of immunology and infectious diseases Iran University of Medical Sciences, Tehran, Iran.
| | - Nazanin Mojtabavi
- Department of Immunology, School of Medicine Iran University of Medical Sciences, Tehran, Iran; Immunology research center institute of immunology and infectious diseases Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Wang B, Golubov J, Oswald EM, Poon P, Wei Q, Lett C, Shehadeh F, Kaczynski M, Felix LO, Mishra B, Mylona EK, Wipperman MF, Chio E, Hamon SC, Hooper AT, Somersan-Karakaya S, Musser BJ, Petro CD, Hamilton JD, Sleeman MA, Kalliolias GD, Mylonakis E, Skokos D. Potential immunomodulatory effects of CAS+IMD monoclonal antibody cocktail in hospitalized patients with COVID-19. EBioMedicine 2024; 108:105334. [PMID: 39270622 PMCID: PMC11415811 DOI: 10.1016/j.ebiom.2024.105334] [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: 01/19/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Passive administration of SARS-CoV-2 neutralizing monoclonal antibodies (mAbs), such as CAS + IMD (Casirivimab + Imdevimab) antibody cocktail demonstrated beneficial effects on clinical outcomes in hospitalized patients with COVID-19 who were seronegative at baseline and outpatients. However, little is known about their impact on the host immunophenotypes. METHODS We conducted an immunoprofiling study in 46 patients from a single site of a multi-site trial of CAS + IMD in hospitalized patients. We collected longitudinal samples during October 2020 ∼ April 2021, prior to the emergence of the Delta and Omicron variants and the use of COVID-19 vaccines. All collected samples were analyzed without exclusion and post-hoc statistical analysis was performed. We examined the dynamic interplay of CAS + IMD with host immunity applying dimensional reduction approach on plasma proteomics and high dimensional flow cytometry data. FINDINGS Using an unbiased clustering method, we identified unique immunophenotypes associated with acute inflammation and disease resolution. Compared to placebo group, administration of CAS + IMD accelerated the transition from an acute inflammatory immunophenotype, to a less inflammatory or "resolving" immunophenotype, as characterized by reduced tissue injury, proinflammatory markers and restored lymphocyte/monocyte imbalance independent of baseline serostatus. Moreover, CAS + IMD did not impair the magnitude or the quality of host T cell immunity against SARS-CoV-2 spike protein. INTERPRETATION Our results identified immunophenotypic changes indicative of a possible SARS-CoV-2 neutralizing antibodies-induced anti-inflammatory effect, without an evident impairment of cellular antiviral immunity, suggesting that further studies of Mabs effects on SAS-CoV-2 or other viral mediated inflammation are warranted. FUNDING Regeneron Pharmaceuticals Inc and federal funds from the Department of Health and Human Services; Administration for Strategic Preparedness and Response; Biomedical Advanced Research and Development Authority, under OT number: HHSO100201700020C.
Collapse
Affiliation(s)
- Bei Wang
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | - Erin M Oswald
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Patrick Poon
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Qiaozhi Wei
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Clarissa Lett
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Fadi Shehadeh
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Matthew Kaczynski
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA
| | - Lewis Oscar Felix
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Biswajit Mishra
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Evangelia K Mylona
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | | | - Erica Chio
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Sara C Hamon
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Andrea T Hooper
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | - Bret J Musser
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | | | - Matthew A Sleeman
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | - Eleftherios Mylonakis
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Dimitris Skokos
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA.
| |
Collapse
|
3
|
Behera N, Patra JK, Dash BK, Pattnaik M, Sahu D, Rambhoopal Reddy B. Clinico-radiological and pulmonary function assessment of post-COVID-19 patients with respiratory symptoms. J Family Med Prim Care 2024; 13:2912-2920. [PMID: 39228580 PMCID: PMC11368303 DOI: 10.4103/jfmpc.jfmpc_1721_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 09/05/2024] Open
Abstract
Background Respiratory symptoms may persist for several weeks following the initial coronavirus disease 2019 (COVID-19) infection. The aims and objectives were to assess the clinical symptoms, pulmonary functions, and radiological changes and to assess the cardio-vascular complications in post-COVID-19 patients. Methods This observational study was conducted in the Department of Pulmonary Medicine in collaboration with the Department of Cardiology, SCBMCH, Cuttack, from March 2021 to August 2022 on 75 post-COVID-19 patients with respiratory symptoms from 4 weeks to 2 years after treatment for COVID-19 infection. Post-COVID patients having previous respiratory diseases were excluded from the study. Results Among 75 patients, the most common age group was 18-30 years with a male-to-female ratio of 2.5:1. Based on O2 requirement, patients were divided into the mild symptomatic group and moderate to severe pneumonia group. The most common respiratory symptom was dyspnea, followed by cough with expectoration. Bilateral crepitations were found in 17% of cases. C-reactive protein (CRP) and D-dimer were increased in 38.6% and 32% of patients, respectively. 42.6% had abnormal chest X-ray, and the most common abnormal finding was reticular thickening. In spirometry, the restrictive pattern and mixed pattern were the predominant types documented in 49.3% and 13.3% of cases, respectively, which were significant in the moderate-severe group. Diffusion capacity of the lungs for carbon monoxide (DLCO) was performed in only 19 patients (mild group 13 and moderate-severe group 6). Twelve (63.2%) patients had abnormal DLCO. P- values were significant for RV (0.0482) and RV/TLC (0.0394). High-resolution computed tomography (HRCT) of the thorax was abnormal in 55.7% with the most common abnormalities as inter- and intra-lobular septal thickening. The left ventricular ejection fraction was preserved in all patients, with right atrium and right ventricle enlargement in 2.6% and pulmonary hypertension in 4.0% of participants. Conclusion All post-COVID-19 patients having respiratory symptoms after recovery from acute COVID-19 may be referred by family care physicians to a dedicated post-COVID center for further evaluation, management, and early rehabilitation to decrease the morbidity in recovered patients. Persistent increased blood parameters like TLC, N/L ratio, RBS, CRP, and D-dimer seen in recovered post-COVID-19 patients. The long-term impact of CT findings on respiratory symptoms, pulmonary functions, and quality of life is unknown. Cardiovascular abnormalities in post-COVID-19 patients are infrequent.
Collapse
Affiliation(s)
- Nilakantha Behera
- Department of Pulmonary Medicine, SCB Medical College and Hospital, Cuttack, Odisha, India
| | - Jeetendra Kumar Patra
- Department of Pulmonary Medicine, SCB Medical College and Hospital, Cuttack, Odisha, India
| | - Bijay Kumar Dash
- Department of Cardiology, SCB Medical College and Hospital, Cuttack, Odisha, India
| | - Manoranjan Pattnaik
- Department of Pulmonary Medicine, SCB Medical College and Hospital, Cuttack, Odisha, India
| | - Deepak Sahu
- Department of Community Medicine, SJ Medical College and Hospital, Puri, Odisha, India
| | | |
Collapse
|
4
|
Hofmann H, Önder A, Becker J, Gröger M, Müller MM, Zink F, Stein B, Radermacher P, Waller C. Markers of oxidative stress during post-COVID-19 fatigue: a hypothesis-generating, exploratory pilot study on hospital employees. Front Med (Lausanne) 2023; 10:1305009. [PMID: 38111693 PMCID: PMC10725950 DOI: 10.3389/fmed.2023.1305009] [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: 09/30/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023] Open
Abstract
Introduction Post-COVID-19 fatigue is common after recovery from COVID-19. Excess formation of reactive oxygen species (ROS) leading to oxidative stress-related mitochondrial dysfunction is referred to as a cause of these chronic fatigue-like symptoms. The present observational pilot study aimed to investigate a possible relationship between the course of ROS formation, subsequent oxidative stress, and post-COVID-19 fatigue. Method A total of 21 post-COVID-19 employees of the General Hospital Nuremberg suffering from fatigue-like symptoms were studied during their first consultation (T1: on average 3 months after recovery from COVID-19), which comprised an educational talk on post-COVID-19 symptomatology and individualized outpatient strategies to resume normal activity, and 8 weeks thereafter (T2). Fatigue severity was quantified using the Chalder Fatigue Scale together with a health survey (Patient Health Questionnaire) and self-report on wellbeing (12-Item Short-Form Health Survey). We measured whole blood superoxide anion (O 2 • - ) production rate (electron spin resonance, as a surrogate for ROS production) and oxidative stress-induced DNA strand breaks (single cell gel electrophoresis: "tail moment" in the "comet assay"). Results Data are presented as mean ± SD or median (interquartile range) depending on the data distribution. Differences between T1 and T2 were tested using a paired Wilcoxon rank sign or t-test. Fatigue intensity decreased from 24 ± 5 at T1 to 18 ± 8 at T2 (p < 0.05), which coincided with reduced O 2 • - formation (from 239 ± 55 to 195 ± 59 nmol/s; p < 0.05) and attenuated DNA damage [tail moment from 0.67 (0.36-1.28) to 0.32 (0.23-0.71); p = 0.05]. Discussion Our pilot study shows that post-COVID-19 fatigue coincides with (i) enhanced O 2 • - formation and oxidative stress, which are (ii) reduced with attenuation of fatigue symptoms.
Collapse
Affiliation(s)
- Hanna Hofmann
- Department of Psychosomatic Medicine and Psychotherapy, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Alexandra Önder
- Department of Psychosomatic Medicine and Psychotherapy, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Juliane Becker
- Department of Psychosomatic Medicine and Psychotherapy, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Michael Gröger
- Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Markus M Müller
- Department of Psychosomatic Medicine and Psychotherapy, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Fabian Zink
- Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Barbara Stein
- Department of Psychosomatic Medicine and Psychotherapy, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| | - Peter Radermacher
- Anesthesiological Pathophysiology and Process Engineering, University Hospital, Ulm, Germany
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, General Hospital Nuremberg, Paracelsus Medical University, Nuremberg, Germany
| |
Collapse
|
5
|
Al Jahdhami I, Arshad H, Omar H, Bennji SM, Al Nomani K, Al Ghafri M, Al Syabi M, Al Hinai S, Al Mawali A. Persistence of Symptoms Following Hospitalization for COVID-19 in Oman:
A Bidirectional Observational Study. Oman Med J 2023; 38:e570. [PMID: 38283209 PMCID: PMC10822130 DOI: 10.5001/omj.2023.120] [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: 03/31/2023] [Accepted: 07/02/2023] [Indexed: 01/30/2024] Open
Abstract
Objectives This study sought to assess the prevalence of persistent COVID-19-related symptoms in patients with mild, severe, and critical disease. Methods We conducted a bidirectional cohort observational study that included all adult patients ≥ 18 years, admitted to Armed Forces Hospital, Muscat between July 2020 and June 2022, with COVID-19 infection and discharged alive. Patients were requested to attend outpatient clinic at weeks six and 12 post-discharge, where they filled out a questionnaire and underwent a chest X-ray. Additionally, blood tests were performed if necessary. Healthcare workers with mild infections were also requested to fill out a questionnaire about their initial symptoms, persistent symptoms, and comorbidities. Results The study included 468 patients, comprising 261 hospitalized patients and 207 healthcare workers. On follow-up, 39.7% of patients presented with residual symptoms, such as cough, breathlessness, and joint pain. These symptoms were more common in patients with medical comorbidities, particularly hypertension, diabetes, and dyslipidemia. Notably, these symptoms were also observed in patients with mild disease. Post-COVID-19 pulmonary fibrosis was observed in 21 patients, mainly among those admitted to the intensive care unit or requiring prolonged hospitalization. Conclusions This study highlights the persistence of symptoms and the prevalence of post-COVID-19 syndrome at two months post-discharge, especially among patients with severe and critical disease during the acute phase. Various predictors of post-COVID-19 syndrome were identified, including female gender, older age, presence of comorbidities, disease severity, and hypertension. Therefore, patients in these categories require thorough evaluation and long-term follow-up to manage residual symptoms.
Collapse
Affiliation(s)
| | - Husna Arshad
- Department of Medicine, Armed Forces Hospital, Muscat, Oman
| | - Hiba Omar
- Endemic Medicine and Hepatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sami M. Bennji
- Sultan Qaboos Comprehensive Cancer Care and Research Centre, Muscat, Oman
| | | | | | | | | | - Adhra Al Mawali
- Department of Quality Assurance and Planning, German University of Technology, Muscat, Oman
| |
Collapse
|
6
|
Kuchler T, Günthner R, Ribeiro A, Hausinger R, Streese L, Wöhnl A, Kesseler V, Negele J, Assali T, Carbajo-Lozoya J, Lech M, Schneider H, Adorjan K, Stubbe HC, Hanssen H, Kotilar K, Haller B, Heemann U, Schmaderer C. Persistent endothelial dysfunction in post-COVID-19 syndrome and its associations with symptom severity and chronic inflammation. Angiogenesis 2023; 26:547-563. [PMID: 37507580 PMCID: PMC10542303 DOI: 10.1007/s10456-023-09885-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Post-COVID-19 syndrome (PCS) is a lingering disease with ongoing symptoms such as fatigue and cognitive impairment resulting in a high impact on the daily life of patients. Understanding the pathophysiology of PCS is a public health priority, as it still poses a diagnostic and treatment challenge for physicians. METHODS In this prospective observational cohort study, we analyzed the retinal microcirculation using Retinal Vessel Analysis (RVA) in a cohort of patients with PCS and compared it to an age- and gender-matched healthy cohort (n = 41, matched out of n = 204). MEASUREMENTS AND MAIN RESULTS PCS patients exhibit persistent endothelial dysfunction (ED), as indicated by significantly lower venular flicker-induced dilation (vFID; 3.42% ± 1.77% vs. 4.64% ± 2.59%; p = 0.02), narrower central retinal artery equivalent (CRAE; 178.1 [167.5-190.2] vs. 189.1 [179.4-197.2], p = 0.01) and lower arteriolar-venular ratio (AVR; (0.84 [0.8-0.9] vs. 0.88 [0.8-0.9], p = 0.007). When combining AVR and vFID, predicted scores reached good ability to discriminate groups (area under the curve: 0.75). Higher PCS severity scores correlated with lower AVR (R = - 0.37 p = 0.017). The association of microvascular changes with PCS severity were amplified in PCS patients exhibiting higher levels of inflammatory parameters. CONCLUSION Our results demonstrate that prolonged endothelial dysfunction is a hallmark of PCS, and impairments of the microcirculation seem to explain ongoing symptoms in patients. As potential therapies for PCS emerge, RVA parameters may become relevant as clinical biomarkers for diagnosis and therapy management. TRIAL REGISTRATION This study was previously registered at ClinicalTrials ("All Eyes on PCS-Analysis of the Retinal Microvasculature in Patients with Post-COVID-19 Syndrome". NCT05635552. https://clinicaltrials.gov/ct2/show/NCT05635552 ). Persistent endothelial dysfunction in post-COVID-19 syndrome. Acute SARS-CoV-2 infection indirectly or directly causes endotheliitis in patients. N = 41 PCS patients were recruited and retinal vessel analysis was performed to assess microvascular endothelial function. Images of SVA and DVA are illustrative for RVA data analysis. For each PCS patient and healthy cohort, venular vessel diameter of the three measurement cycles was calculated and plotted on a diameter-time curve. Patients exhibited reduced flicker-induced dilation in veins (vFID) measured by dynamic vessel analysis (DVA) and lower central retinal arteriolar equivalent (CRAE) and arteriolar-venular ratio (AVR) and a tendency towards higher central retinal venular equivalent (CRVE) when compared to SARS-CoV-2 infection naïve participants. Created with BioRender.com.
Collapse
Affiliation(s)
- Timon Kuchler
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Roman Günthner
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Andrea Ribeiro
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
- Medizinische Klinik Und Poliklinik IV, LMU University Hospital Munich, Ziemssenstraße 5, 80336, Munich, Germany
| | - Renate Hausinger
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Lukas Streese
- Faculty of Health Care, Niederrhein University of Applied Sciences, Krefeld, Germany
| | - Anna Wöhnl
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Veronika Kesseler
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Johanna Negele
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Tarek Assali
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Javier Carbajo-Lozoya
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Maciej Lech
- Medizinische Klinik Und Poliklinik IV, LMU University Hospital Munich, Ziemssenstraße 5, 80336, Munich, Germany
| | - Heike Schneider
- School of Medicine, Klinikum Rechts Der Isar, Department of Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Kristina Adorjan
- Department of Psychiatry and Psychotherapy, LMU University Hospital Munich, Nußbaumstraße 7, 80336, Munich, Germany
| | - Hans Christian Stubbe
- Medizinische Klinik Und Poliklinik II, LMU University Hospital Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Henner Hanssen
- Department of Sport, Exercise and Health, Preventive Sports Medicine and Systems Physiology, University of Basel, Basel, Switzerland
| | - Konstantin Kotilar
- Aachen University of Applied Sciences, Heinrich-Mussmann-Str. 1, 52428, Jülich, Germany
| | - Bernhard Haller
- School of Medicine, Institute for AI and Informatics in Medicine, Technical University of Munich, Klinikum Rechts Der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Uwe Heemann
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Christoph Schmaderer
- School of Medicine, Klinikum Rechts Der Isar, Department of Nephrology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.
- German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
7
|
Korobova ZR, Arsentieva NA, Totolian AA. Macrophage-Derived Chemokine MDC/CCL22: An Ambiguous Finding in COVID-19. Int J Mol Sci 2023; 24:13083. [PMID: 37685890 PMCID: PMC10487728 DOI: 10.3390/ijms241713083] [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: 07/13/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Macrophage-derived chemokine (MDC/CCL22) is a chemokine of the C-C subfamily. It is involved in T-cellular maturation and migration. Our previous research shows that plasma CCL22/MDC tends to show a statistically significant depletion of concentrations in acute patients and convalescents when compared to healthy donors. In the current work, we investigate existing views on MDC/CCL22 dynamics in association with various pathologies, including respiratory diseases and, specifically, COVID-19. Additionally, we present our explanations for the observed decrease in MDC/CCL22 concentrations in COVID-19. The first hypothesis we provide implies that viral products bind to MDC/CCL22 and block its activity. Another explanation for this phenomenon is based on dendritic cells population and the inhibition of their function.
Collapse
Affiliation(s)
- Zoia R. Korobova
- Laboratory of Molecular Immunology, Saint Petersburg Pasteur Institute, Mira St. 14, 197101 St. Petersburg, Russia; (Z.R.K.); (N.A.A.)
- Department of Immunology, Pavlov First State Medical University of St. Petersburg, L’va Tolstogo St. 6–8, 197022 St. Petersburg, Russia
| | - Natalia A. Arsentieva
- Laboratory of Molecular Immunology, Saint Petersburg Pasteur Institute, Mira St. 14, 197101 St. Petersburg, Russia; (Z.R.K.); (N.A.A.)
- Department of Immunology, Pavlov First State Medical University of St. Petersburg, L’va Tolstogo St. 6–8, 197022 St. Petersburg, Russia
| | - Areg A. Totolian
- Laboratory of Molecular Immunology, Saint Petersburg Pasteur Institute, Mira St. 14, 197101 St. Petersburg, Russia; (Z.R.K.); (N.A.A.)
- Department of Immunology, Pavlov First State Medical University of St. Petersburg, L’va Tolstogo St. 6–8, 197022 St. Petersburg, Russia
| |
Collapse
|
8
|
Wiethoff JP, Sandmann S, Theiler T, Nze Nkogue C, Akomo-Okoue EF, Varghese J, Kreidenweiss A, Mellmann A, Lell B, Adegnika AA, Held J, Schaumburg F. Pharyngeal Communities and Antimicrobial Resistance in Pangolins in Gabon. Microbiol Spectr 2023; 11:e0066423. [PMID: 37338382 PMCID: PMC10434165 DOI: 10.1128/spectrum.00664-23] [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: 02/13/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
Wildlife can be a reservoir and source of zoonotic pathogens for humans. For instance, pangolins were considered one of the potential animal reservoirs of SARS-CoV-2. The aim of this study was to assess the prevalence of antimicrobial-resistant species (e.g., extended-spectrum β-lactamase [ESBL]-producing Enterobacterales) and Staphylococcus aureus-related complex and to describe the bacterial community in wild Gabonese pangolins. The pharyngeal colonization of pangolins sold in Gabon (n = 89, 2021 to 2022) was analyzed using culture media selective for ESBL-producing Enterobacterales, S. aureus-related complex, Gram-positive bacteria and nonfermenters. Phylogenetic analyses of ESBL-producing Enterobacterales was done using core-genome multilocus sequence typing (cgMLST) and compared with publicly available genomes. Patterns of cooccurring species were detected by network analysis. Of the 439 bacterial isolates, the majority of species belonged to the genus Pseudomonas (n = 170), followed by Stenotrophomonas (n = 113) and Achromobacter (n = 37). Three Klebsiella pneumoniae isolates and one Escherichia coli isolate were ESBL-producers, which clustered with human isolates from Nigeria (MLST sequence type 1788 [ST1788]) and Gabon (ST38), respectively. Network analysis revealed a frequent cooccurrence of Stenotrophomonas maltophilia with Pseudomonas putida and Pseudomonas aeruginosa. In conclusion, pangolins can be colonized with human-related ESBL-producing K. pneumoniae and E. coli. Unlike in other African wildlife, S. aureus-related complex was not detected in pangolins. IMPORTANCE There is an ongoing debate if pangolins are a relevant reservoir for viruses such as SARS-CoV-2. Here, we wanted to know if African pangolins are colonized with bacteria that are relevant for human health. A wildlife reservoir of antimicrobial resistance would be of medical relevance in regions were consumption of so-called bushmeat is common. In 89 pangolins, we found three ESBL-producing Klebsiella pneumoniae strains and one ESBL-producing Escherichia coli strains, which were closely related to isolates from humans in Africa. This points toward either a transmission between pangolins and humans or a common source from which both humans and pangolins became colonized.
Collapse
Affiliation(s)
- Johanna P. Wiethoff
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Tom Theiler
- Institute of Medical Microbiology, University of Münster, Münster, Germany
| | | | | | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
| | | | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Ayôla A. Adegnika
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Frieder Schaumburg
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Medical Microbiology, University of Münster, Münster, Germany
| |
Collapse
|
9
|
Tsiakalos A, Ziakas PD, Polyzou E, Schinas G, Akinosoglou K. Early Fluvoxamine Reduces the Risk for Clinical Deterioration in Symptomatic Outpatients with COVID-19: A Real-World, Retrospective, before-after Analysis. Microorganisms 2023; 11:2073. [PMID: 37630633 PMCID: PMC10459506 DOI: 10.3390/microorganisms11082073] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Fluvoxamine, a selective serotonin reuptake inhibitor with anti-inflammatory properties, has gained attention as a repurposed drug to treat COVID-19. We aimed to explore the potential benefit of fluvoxamine on outpatients with early SARS-CoV-2 infection. We performed a retrospective study of fluvoxamine adult outpatients with symptomatic COVID-19 disease of early onset (<5 days), in the context of an infectious diseases private practice, between September-December 2021, in Greece. Patients with disease duration ≥5 days, dyspnea and/or hypoxemia with oxygen saturation <94% in room air and pregnancy were excluded from the analysis. In total, 103 patients, 54 males/49 females with a median age of 47 years (39-56), were included in this study. Patient characteristics were balanced before and after the introduction of fluvoxamine. Two patients in the fluvoxamine arm (3.8%; 95% CI 0.4-13) had clinical deterioration compared to 8 patients in the standard of care group (16%; 95% CI 7.2-29.1, p < 0.04). After controlling for age, sex, body mass index > 30 and vaccination status, fluvoxamine was independently associated with a lower risk of clinical deterioration (adj. OR 0.12; 95% CI 0.02-0.70, p < 0.02). Adding on fluvoxamine to treatment for early symptomatic COVID-19 patients may protect them from clinical deterioration and hospitalization, and it is an appealing low-cost, low-toxicity option in the community setting and warrants further investigation.
Collapse
Affiliation(s)
| | | | - Eleni Polyzou
- Dept of Internal Medicine and Infectious Diseases, Medical School, University General Hospital of Patras, University of Patras, 26504 Rio, Greece; (E.P.); (G.S.); (K.A.)
| | - Georgios Schinas
- Dept of Internal Medicine and Infectious Diseases, Medical School, University General Hospital of Patras, University of Patras, 26504 Rio, Greece; (E.P.); (G.S.); (K.A.)
| | - Karolina Akinosoglou
- Dept of Internal Medicine and Infectious Diseases, Medical School, University General Hospital of Patras, University of Patras, 26504 Rio, Greece; (E.P.); (G.S.); (K.A.)
| |
Collapse
|
10
|
Ayasoufi K, Wolf DM, Namen SL, Jin F, Tritz ZP, Pfaller CK, Zheng J, Goddery EN, Fain CE, Gulbicki LR, Borchers AL, Reesman RA, Yokanovich LT, Maynes MA, Bamkole MA, Khadka RH, Hansen MJ, Wu LJ, Johnson AJ. Brain resident memory T cells rapidly expand and initiate neuroinflammatory responses following CNS viral infection. Brain Behav Immun 2023; 112:51-76. [PMID: 37236326 PMCID: PMC10527492 DOI: 10.1016/j.bbi.2023.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/25/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
The contribution of circulating verses tissue resident memory T cells (TRMs) to clinical neuropathology is an enduring question due to a lack of mechanistic insights. The prevailing view is TRMs are protective against pathogens in the brain. However, the extent to which antigen-specific TRMs induce neuropathology upon reactivation is understudied. Using the described phenotype of TRMs, we found that brains of naïve mice harbor populations of CD69+ CD103- T cells. Notably, numbers of CD69+ CD103- TRMs rapidly increase following neurological insults of various origins. This TRM expansion precedes infiltration of virus antigen-specific CD8 T cells and is due to proliferation of T cells within the brain. We next evaluated the capacity of antigen-specific TRMs in the brain to induce significant neuroinflammation post virus clearance, including infiltration of inflammatory myeloid cells, activation of T cells in the brain, microglial activation, and significant blood brain barrier disruption. These neuroinflammatory events were induced by TRMs, as depletion of peripheral T cells or blocking T cell trafficking using FTY720 did not change the neuroinflammatory course. Depletion of all CD8 T cells, however, completely abrogated the neuroinflammatory response. Reactivation of antigen-specific TRMs in the brain also induced profound lymphopenia within the blood compartment. We have therefore determined that antigen-specific TRMs can induce significant neuroinflammation, neuropathology, and peripheral immunosuppression. The use of cognate antigen to reactivate CD8 TRMs enables us to isolate the neuropathologic effects induced by this cell type independently of other branches of immunological memory, differentiating this work from studies employing whole pathogen re-challenge. This study also demonstrates the capacity for CD8 TRMs to contribute to pathology associated with neurodegenerative disorders and long-term complications associated with viral infections. Understanding functions of brain TRMs is crucial in investigating their role in neurodegenerative disorders including MS, CNS cancers, and long-term complications associated with viral infections including COVID-19.
Collapse
Affiliation(s)
| | - Delaney M Wolf
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Shelby L Namen
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Zachariah P Tritz
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Christian K Pfaller
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, United States; Paul-Ehrlich-Institut, Langen, Germany
| | - Jiaying Zheng
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Emma N Goddery
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Cori E Fain
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | | | - Anna L Borchers
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | | | - Lila T Yokanovich
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Mark A Maynes
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Michael A Bamkole
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Roman H Khadka
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Michael J Hansen
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Long-Jun Wu
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Molecular Medicine, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States.
| |
Collapse
|
11
|
Tsilingiris D, Vallianou NG, Karampela I, Christodoulatos GS, Papavasileiou G, Petropoulou D, Magkos F, Dalamaga M. Laboratory Findings and Biomarkers in Long COVID: What Do We Know So Far? Insights into Epidemiology, Pathogenesis, Therapeutic Perspectives and Challenges. Int J Mol Sci 2023; 24:10458. [PMID: 37445634 PMCID: PMC10341908 DOI: 10.3390/ijms241310458] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Long COVID (LC) encompasses a constellation of long-term symptoms experienced by at least 10% of people after the initial SARS-CoV-2 infection, and so far it has affected about 65 million people. The etiology of LC remains unclear; however, many pathophysiological pathways may be involved, including viral persistence; a chronic, low-grade inflammatory response; immune dysregulation and a defective immune response; the reactivation of latent viruses; autoimmunity; persistent endothelial dysfunction and coagulopathy; gut dysbiosis; hormonal and metabolic dysregulation; mitochondrial dysfunction; and autonomic nervous system dysfunction. There are no specific tests for the diagnosis of LC, and clinical features including laboratory findings and biomarkers may not specifically relate to LC. Therefore, it is of paramount importance to develop and validate biomarkers that can be employed for the prediction, diagnosis and prognosis of LC and its therapeutic response, although this effort may be hampered by challenges pertaining to the non-specific nature of the majority of clinical manifestations in the LC spectrum, small sample sizes of relevant studies and other methodological issues. Promising candidate biomarkers that are found in some patients are markers of systemic inflammation, including acute phase proteins, cytokines and chemokines; biomarkers reflecting SARS-CoV-2 persistence, the reactivation of herpesviruses and immune dysregulation; biomarkers of endotheliopathy, coagulation and fibrinolysis; microbiota alterations; diverse proteins and metabolites; hormonal and metabolic biomarkers; and cerebrospinal fluid biomarkers. At present, there are only two reviews summarizing relevant biomarkers; however, they do not cover the entire umbrella of current biomarkers, their link to etiopathogenetic mechanisms or the diagnostic work-up in a comprehensive manner. Herein, we aim to appraise and synopsize the available evidence on the typical laboratory manifestations and candidate biomarkers of LC, their classification based on pathogenetic mechanisms and the main LC symptomatology in the frame of the epidemiological and clinical aspects of the syndrome and furthermore assess limitations and challenges as well as potential implications in candidate therapeutic interventions.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece;
| | - Natalia G. Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Street, 10676 Athens, Greece;
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Street, 12462 Athens, Greece;
| | | | - Georgios Papavasileiou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| | - Dimitra Petropoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| | - Faidon Magkos
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, DK-2200 Frederiksberg, Denmark;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| |
Collapse
|
12
|
Sandmann S, Nunes JV, Grobusch MP, Sesay M, Kriegel MA, Varghese J, Schaumburg F. Research article network analysis of polymicrobial chronic wound infections in Masanga, Sierra Leone. BMC Infect Dis 2023; 23:250. [PMID: 37072717 PMCID: PMC10112320 DOI: 10.1186/s12879-023-08204-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/28/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Chronic wounds are frequently colonized or infected with multiple bacterial or fungal species, which can both promote or inhibit each other. Network analyses are helpful to understand the interplay of these species in polymicrobial infections. Our aim was to analyse the network of bacterial and fungal species in chronic wounds. METHODS Swabs (n = 163) from chronic wound infections (Masanga, Sierra Leone, 2019-2020) were screened for bacterial and fungal species using non-selective agars. Some of these wounds were suspected but not confirmed Buruli ulcer. Species identification was done with MALDI-TOF mass spectrometry. Network analysis was performed to investigate co-occurrence of different species within one patient. All species with n ≥ 10 isolates were taken into account. RESULTS Of the 163 patients, 156 had a positive wound culture (median of three different species per patient; range 1-7). Pseudomonas aeruginosa (n = 75) was the dominating species with frequent co-detections of Klebsiella pneumoniae (21 cases; OR = 1.36, 95%CI: 0.63-2.96, p = 0.47), Staphylococcus aureus (14 cases; OR = 1.06, 95%CI: 0.44-2.55, p = 1) and Proteus mirabilis (13 cases; OR = 0.84, 95%CI: 0.35-1.99, p = 0.69). CONCLUSION The culturome of chronic wounds in Sierra Leonean patients is highly diverse and characterized by the co-occurrence of P. aeruginosa, K. pneumoniae and S. aureus.
Collapse
Affiliation(s)
- Sarah Sandmann
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Jonathan Vas Nunes
- Masanga Medical Research Unit (MMRU), Masanga, Sierra Leone
- Department of Infectious Diseases, Center of Tropical Medicine and Travel Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Martin P Grobusch
- Masanga Medical Research Unit (MMRU), Masanga, Sierra Leone
- Department of Infectious Diseases, Center of Tropical Medicine and Travel Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Maxwell Sesay
- Masanga Medical Research Unit (MMRU), Masanga, Sierra Leone
| | - Martin A Kriegel
- Section of Rheumatology and Clinical Immunology, Department of Medicine, University Hospital Münster, Münster, Germany
- Department of Translational Rheumatology and Immunology, Institute of Musculoskeletal Medicine, University of Münster, Münster, Germany
- Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany
- Department of Immunobiology, Yale University School of Medicine, New Haven, USA
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University of Münster, Münster, Germany.
| |
Collapse
|
13
|
Islam MS, Wang Z, Abdel-Mohsen M, Chen X, Montaner LJ. Tissue injury and leukocyte changes in post-acute sequelae of SARS-CoV-2: review of 2833 post-acute patient outcomes per immune dysregulation and microbial translocation in long COVID. J Leukoc Biol 2023; 113:236-254. [PMID: 36807444 DOI: 10.1093/jleuko/qiac001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Indexed: 01/18/2023] Open
Abstract
A significant number of persons with coronavirus disease 2019 (COVID-19) experience persistent, recurrent, or new symptoms several months after the acute stage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This phenomenon, termed post-acute sequelae of SARS-CoV-2 (PASC) or long COVID, is associated with high viral titers during acute infection, a persistently hyperactivated immune system, tissue injury by NETosis-induced micro-thrombofibrosis (NETinjury), microbial translocation, complement deposition, fibrotic macrophages, the presence of autoantibodies, and lymphopenic immune environments. Here, we review the current literature on the immunological imbalances that occur during PASC. Specifically, we focus on data supporting common immunopathogenesis and tissue injury mechanisms shared across this highly heterogenous disorder, including NETosis, coagulopathy, and fibrosis. Mechanisms include changes in leukocyte subsets/functions, fibroblast activation, cytokine imbalances, lower cortisol, autoantibodies, co-pathogen reactivation, and residual immune activation driven by persistent viral antigens and/or microbial translocation. Taken together, we develop the premise that SARS-CoV-2 infection results in PASC as a consequence of acute and/or persistent single or multiple organ injury mediated by PASC determinants to include the degree of host responses (inflammation, NETinjury), residual viral antigen (persistent antigen), and exogenous factors (microbial translocation). Determinants of PASC may be amplified by comorbidities, age, and sex.
Collapse
Affiliation(s)
- Md Sahidul Islam
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China
| | - Zhaoxiong Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China
| | - Mohamed Abdel-Mohsen
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, United States
| | - Xin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China.,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Research Building N22, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China
| | - Luis J Montaner
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, United States
| |
Collapse
|
14
|
Benitez G, Shehadeh F, Mylona EK, Tran QL, Tsikala-Vafea M, Atalla E, Kaczynski M, Mylonakis E. Effect of Thymalfasin (Thymosin-α-1) on Reversing Lymphocytopenia among Patients with COVID-19. Int Immunopharmacol 2023:109831. [PMCID: PMC9902288 DOI: 10.1016/j.intimp.2023.109831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Introduction Thymosin-α-1 (Tα1) elevates lymphocyte counts among patients with COVID-19, but its effect on reversing lymphocytopenia is unknown. Methods 24 patients treated with Tα1 and 100 patients in the control arm were included in this analysis. The incidence rate of reversing lymphocytopenia, overall and stratified by baseline oxygen support, above the threshold for classification of lymphocytopenia (i.e., Total Lymphocyte Count (TLC) < 1.5 x 109/L) and severe lymphocytopenia (i.e., TLC < 1.0 x 109/L) within 3, 5, and 7 days of treatment initiation was calculated, along with incidence rate ratios (IRRs) and 95% confidence intervals (CIs). Results Compared with the standard of care, the rate of reversing lymphocytopenia (IRR: 2.38, 95% CI: 0.92 – 5.81) and severe lymphocytopenia (IRR: 1.57, 95% CI: 0.59 – 3.72), especially among patients with severe lymphocytopenia on high flow oxygen support (IRR: 3.64, 95% CI: 0.71 – 23.44), was greater for patients treated with Tα1 within 3 days of treatment initiation, although analyses were not significant. Conclusion Among patients with hypoxemia and lymphocytopenia, Tα1 may reverse lymphocytopenia and severe lymphocytopenia, particularly within 3 days of treatment initiation, faster than the standard of care.
Collapse
Affiliation(s)
- Gregorio Benitez
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Fadi Shehadeh
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Evangelia K. Mylona
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Quynh-Lam Tran
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Maria Tsikala-Vafea
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Eleftheria Atalla
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Matthew Kaczynski
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Corresponding author at: Eleftherios Mylonakis 593 Eddy Street, POB, 3rd Floor, Suite 328/330, Providence, RI 02903, USA
| |
Collapse
|
15
|
Dufrusine B, Valentinuzzi S, Bibbò S, Damiani V, Lanuti P, Pieragostino D, Del Boccio P, D’Alessandro E, Rabottini A, Berghella A, Allocati N, Falasca K, Ucciferri C, Mucedola F, Di Perna M, Martino L, Vecchiet J, De Laurenzi V, Dainese E. Iron Dyshomeostasis in COVID-19: Biomarkers Reveal a Functional Link to 5-Lipoxygenase Activation. Int J Mol Sci 2022; 24:15. [PMID: 36613462 PMCID: PMC9819889 DOI: 10.3390/ijms24010015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is characterized by a broad spectrum of clinical symptoms. After acute infection, some subjects develop a post-COVID-19 syndrome known as long-COVID. This study aims to recognize the molecular and functional mechanisms that occur in COVID-19 and long-COVID patients and identify useful biomarkers for the management of patients with COVID-19 and long-COVID. Here, we profiled the response to COVID-19 by performing a proteomic analysis of lymphocytes isolated from patients. We identified significant changes in proteins involved in iron metabolism using different biochemical analyses, considering ceruloplasmin (Cp), transferrin (Tf), hemopexin (HPX), lipocalin 2 (LCN2), and superoxide dismutase 1 (SOD1). Moreover, our results show an activation of 5-lipoxygenase (5-LOX) in COVID-19 and in long-COVID possibly through an iron-dependent post-translational mechanism. Furthermore, this work defines leukotriene B4 (LTB4) and lipocalin 2 (LCN2) as possible markers of COVID-19 and long-COVID and suggests novel opportunities for prevention and treatment.
Collapse
Affiliation(s)
- Beatrice Dufrusine
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Silvia Valentinuzzi
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Sandra Bibbò
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Verena Damiani
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Ersilia D’Alessandro
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Alberto Rabottini
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Alessandro Berghella
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nerino Allocati
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Katia Falasca
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Claudio Ucciferri
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Francesco Mucedola
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Marco Di Perna
- Pneumology Department, “SS Annunziata” Hospital, 66100 Chieti, Italy
| | - Laura Martino
- Pneumology Department, “SS Annunziata” Hospital, 66100 Chieti, Italy
| | - Jacopo Vecchiet
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Enrico Dainese
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
16
|
Swarnakar R, Jenifa S, Wadhwa S. Musculoskeletal complications in long COVID-19: A systematic review. World J Virol 2022; 11:485-495. [PMID: 36483107 PMCID: PMC9724204 DOI: 10.5501/wjv.v11.i6.485] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has crippled humanity since early 2020. Various sequelae of COVID-19 have been reported in different body systems. Musculoskeletal symptoms are widely reported during COVID-19 infection, but musculoskeletal complications in long COVID-19 are underreported. However, post-COVID-19 survivors have reported complaints of persisting or new-onset fatigue, myalgia, arthralgia, arthritis, muscle weakness, etc in clinical practice. The well-known detrimental effects of steroids on the musculoskeletal system coupled with their over-the-counter availability can also be anticipated since they were the cornerstone of life-saving management in this pandemic.
AIM To determine the musculoskeletal complications in long COVID.
METHODS We performed a systematic review of ‘systematic reviews and meta-analyses’.
RESULTS Of the 63 articles screened, 24 articles were included. Two articles specifically discussed children and adolescents. One article discussed rehabilitation intervention. No article addressed rehabilitation of musculoskeletal issues in long COVID-19 in particular. Fatigue was the most common musculoskeletal complication.
CONCLUSION Fatigue is found to be very common along with myalgia and arthralgia. There were no studies on rehabilitation intervention in musculoskeletal complications specifically. Considering the lacuna in literature and the needs of the current situation, further studies are warranted to standardize effective rehabilitation interventions in musculoskeletal complications. More homogenous studies are needed. Studies on functional impairment due to musculoskeletal involvement are essential.
Collapse
Affiliation(s)
- Raktim Swarnakar
- Department of Physical Medicine and Rehabilitation, All India Institute of Medical Sciences, New Delhi 110029, Delhi, India
| | - Shoibam Jenifa
- Department of Physical Medicine and Rehabilitation, All India Institute of Medical Sciences, New Delhi 110029, Delhi, India
| | - Sanjay Wadhwa
- Department of Physical Medicine and Rehabilitation, All India Institute of Medical Sciences, New Delhi 110029, Delhi, India
| |
Collapse
|
17
|
Zheng B, Daines L, Han Q, Hurst JR, Pfeffer P, Shankar-Hari M, Elneima O, Walker S, Brown JS, Siddiqui S, Quint JK, Brightling CE, Evans RA, Wain LV, Heaney LG, Sheikh A. Prevalence, risk factors and treatments for post-COVID-19 breathlessness: a systematic review and meta-analysis. Eur Respir Rev 2022; 31:31/166/220071. [PMID: 36323418 PMCID: PMC9724798 DOI: 10.1183/16000617.0071-2022] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/04/2022] [Indexed: 11/05/2022] Open
Abstract
Persistent breathlessness >28 days after acute COVID-19 infection has been identified as a highly debilitating post-COVID symptom. However, the prevalence, risk factors, mechanisms and treatments for post-COVID breathlessness remain poorly understood. We systematically searched PubMed and Embase for relevant studies published from 1 January 2020 to 1 November 2021 (PROSPERO registration number: CRD42021285733) and included 119 eligible papers. Random-effects meta-analysis of 42 872 patients with COVID-19 reported in 102 papers found an overall prevalence of post-COVID breathlessness of 26% (95% CI 23-29) when measuring the presence/absence of the symptom, and 41% (95% CI 34-48) when using Medical Research Council (MRC)/modified MRC dyspnoea scale. The pooled prevalence decreased significantly from 1-6 months to 7-12 months post-infection. Post-COVID breathlessness was more common in those with severe/critical acute infection, those who were hospitalised and females, and was less likely to be reported by patients in Asia than those in Europe or North America. Multiple pathophysiological mechanisms have been proposed (including deconditioning, restrictive/obstructive airflow limitation, systemic inflammation, impaired mental health), but the body of evidence remains inconclusive. Seven cohort studies and one randomised controlled trial suggested rehabilitation exercises may reduce post-COVID breathlessness. There is an urgent need for mechanistic research and development of interventions for the prevention and treatment of post-COVID breathlessness.
Collapse
Affiliation(s)
- Bang Zheng
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Luke Daines
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Qing Han
- Dept of Social Policy and Intervention, University of Oxford, Oxford, UK
| | - John R. Hurst
- UCL Respiratory, University College London, London, UK
| | - Paul Pfeffer
- Barts Health NHS Trust, London, UK,Queen Mary University of London, London, UK
| | - Manu Shankar-Hari
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Omer Elneima
- Institute for Lung Health, Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, UK
| | | | | | - Salman Siddiqui
- Institute for Lung Health, Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, UK,Dept of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Jennifer K. Quint
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Christopher E. Brightling
- Institute for Lung Health, Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Rachael A. Evans
- Institute for Lung Health, Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Louise V. Wain
- Institute for Lung Health, Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, UK,Dept of Health Sciences, University of Leicester, Leicester, UK
| | - Liam G. Heaney
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Aziz Sheikh
- Usher Institute, University of Edinburgh, Edinburgh, UK,Corresponding author: Aziz Sheikh ()
| |
Collapse
|
18
|
Batiha GES, Al-kuraishy HM, Al-Gareeb AI, Welson NN. Pathophysiology of Post-COVID syndromes: a new perspective. Virol J 2022; 19:158. [PMID: 36210445 PMCID: PMC9548310 DOI: 10.1186/s12985-022-01891-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Most COVID-19 patients recovered with low mortality; however, some patients experienced long-term symptoms described as "long-COVID" or "Post-COVID syndrome" (PCS). Patients may have persisting symptoms for weeks after acute SARS-CoV-2 infection, including dyspnea, fatigue, myalgia, insomnia, cognitive and olfactory disorders. These symptoms may last for months in some patients. PCS may progress in association with the development of mast cell activation syndrome (MCAS), which is a distinct kind of mast cell activation disorder, characterized by hyper-activation of mast cells with inappropriate and excessive release of chemical mediators. COVID-19 survivors, mainly women, and patients with persistent severe fatigue for 10 weeks after recovery with a history of neuropsychiatric disorders are more prone to develop PCS. High D-dimer levels and blood urea nitrogen were observed to be risk factors associated with pulmonary dysfunction in COVID-19 survivors 3 months post-hospital discharge with the development of PCS. PCS has systemic manifestations that resolve with time with no further complications. However, the final outcomes of PCS are chiefly unknown. Persistence of inflammatory reactions, autoimmune mimicry, and reactivation of pathogens together with host microbiome alterations may contribute to the development of PCS. The deregulated release of inflammatory mediators in MCAS produces extraordinary symptoms in patients with PCS. The development of MCAS during the course of SARS-CoV-2 infection is correlated to COVID-19 severity and the development of PCS. Therefore, MCAS is treated by antihistamines, inhibition of synthesis of mediators, inhibition of mediator release, and inhibition of degranulation of mast cells.
Collapse
Affiliation(s)
- Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Al Beheira, 22511 Egypt
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Nermeen N. Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni Suef, 62511 Egypt
| |
Collapse
|
19
|
Kimmig LM, Rako ZA, Ziegler S, Richter MJ, G S AT, Roller F, Grimminger F, Vadász I, Seeger W, Herold S, Tello K, Matt U. Long-term comprehensive cardiopulmonary phenotyping of COVID-19. Respir Res 2022; 23:263. [PMID: 36131349 PMCID: PMC9491263 DOI: 10.1186/s12931-022-02173-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
Background Persistent symptoms after initial COVID-19 infection are common and are frequently referred to by the umbrella terms “post-COVID syndrome” and “long COVID”. The sheer number of affected patients pose an increasing challenge to healthcare systems worldwide. To date, our understanding of the pathophysiology of the post-COVID syndrome remains poor and the extent to which persistent cardiopulmonary abnormalities contribute to the symptom complex is unclear. We sought to determine the presence and impact of cardiopulmonary sequelae after COVID-19 in longitudinal assessment. Methods We report on 71 patients who underwent comprehensive, longitudinal testing in regular intervals for up to 12 months after their initial COVID-19 diagnosis. Testing included pulmonary function testing, cardiopulmonary exercise testing, dedicated left and right heart echocardiography, lung ultrasonography, and cardiac MRI. Results Our results demonstrate that subjective quality of life after COVID-19 (EQ-5D visual acuity scale, VAS, 67.4 for patients treated as outpatient, 79.2 for patients admitted to the general floor, 71.8 for patients treated in an ICU) is not related to the severity of the initial infection. Maximal exercise capacity is also reduced (VO2max 79% predicted, SD ± 19%); however, this is driven in large parts by patients who had initially required ICU-level of care. The degree of objective reduction in exertion did not correlate with quality of life scores. Pulmonary function testing revealed mild and persistent reduction in DLCO over the first 12 months without significant restrictive or obstructive lung disease. Left and right heart function was intact with good RV function and intact RV/PA coupling, imaging findings suggestive of myocarditis were uncommon (7% of patients). Conclusion A reduction in exercise capacity after COVID-19 is common, but is most prominent in patients previously treated in the ICU and more likely related to deconditioning or fatigue than to cardiopulmonary impairment. Subjective quality of life scores are independent of the severity of initial infection and do not correlate with objective measures of cardiopulmonary function. In our cohort, persistent cardiopulmonary impairment after COVID-19 was uncommon. The post-COVID syndrome is unlikely to be the result of cardiopulmonary sequalae and may reflect a post-ICU syndrome in some. Trial registration Registered on clinicaltrials.gov (NCT04442789), Date: June 23, 2020 Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02173-9.
Collapse
Affiliation(s)
- Lucas M Kimmig
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany. .,Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA.
| | - Zvonimir A Rako
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefanie Ziegler
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Manuel J Richter
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ashkan Tolou G S
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Fritz Roller
- Department of Radiology, University Hospital Giessen, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany.,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Susanne Herold
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany.,Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Khodr Tello
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ulrich Matt
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Member of the German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
20
|
Shehadeh F, Benitez G, Mylona EK, Tran QL, Tsikala-Vafea M, Atalla E, Kaczynski M, Mylonakis E. A Pilot Trial of Thymalfasin (Thymosin-α-1) to Treat Hospitalized Patients With Hypoxemia and Lymphocytopenia Due to Coronavirus Disease 2019 Infection. J Infect Dis 2022; 227:226-235. [PMID: 36056913 PMCID: PMC9494344 DOI: 10.1093/infdis/jiac362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Thymosin-α-1 (Tα1) may be a treatment option for coronavirus disease 2019 (COVID-19), but efficacy and safety data remain limited. METHODS Prospective, open-label, randomized trial assessing preliminary efficacy and safety of thymalfasin (synthetic form of Tα1), compared with the standard of care, among hospitalized patients with hypoxemia and lymphocytopenia due to COVID-19. RESULTS A total of 49 patients were included in this analysis. Compared with control patients, the incidence of clinical recovery was higher for treated patients with either baseline low-flow oxygen (subdistribution hazard ratio, 1.48 [95% confidence interval, .68-3.25]) or baseline high-flow oxygen (1.28 [.35-4.63]), although neither difference was significant. Among patients with baseline low-flow oxygen, treated patients, compared with control patients, had an average difference of 3.84 times more CD4+ T cells on day 5 than on day 1 (P = .01). Nine serious adverse events among treated patients were deemed not related to Tα1. CONCLUSIONS Tα1 increases CD4+ T-cell count among patients with baseline low-flow oxygen support faster than the standard of care and may have a role in the management of hospitalized patients with hypoxemia and lymphocytopenia due to COVID-19. CLINICAL TRIALS REGISTRATION NCT04487444.
Collapse
Affiliation(s)
| | | | - Evangelia K Mylona
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Quynh Lam Tran
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Maria Tsikala-Vafea
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eleftheria Atalla
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Matthew Kaczynski
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Eleftherios Mylonakis
- Correspondence to: Eleftherios Mylonakis 593 Eddy Street, POB, 3rd Floor, Suite 328/330, Providence, RI 02903, USA.
| |
Collapse
|
21
|
Koumpias AM, Schwartzman D, Fleming O. Long-haul COVID: healthcare utilization and medical expenditures 6 months post-diagnosis. BMC Health Serv Res 2022; 22:1010. [PMID: 35941617 PMCID: PMC9358916 DOI: 10.1186/s12913-022-08387-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Despite extensive evidence that COVID-19 symptoms may persist for up to a year, their long-term implications for healthcare utilization and costs 6 months post-diagnosis remain relatively unexplored. We examine patient-level association of COVID-19 diagnosis association of COVID-19 diagnosis with average monthly healthcare utilization and medical expenditures for up to 6 months, explore heterogeneity across age groups and determine for how many months post-diagnosis healthcare utilization and costs of COVID-19 patients persist above pre-diagnosis levels. METHODS This population-based retrospective cohort study followed COVID-19 patients' healthcare utilization and costs from January 2019 through March 2021 using claims data provided by the COVID-19 Research Database. The patient population includes 250,514 individuals infected with COVID-19 during March-September 2020 and whose last recorded claim was not hospitalization with severe symptoms. We measure the monthly number and costs of total visits and by telemedicine, preventive, urgent care, emergency, immunization, cardiology, inpatient or surgical services and established patient or new patient visits. RESULTS The mean (SD) total number of monthly visits and costs pre-diagnosis were .4783 (4.0839) and 128.06 (1182.78) dollars compared with 1.2078 (8.4962) visits and 351.67 (2473.63) dollars post-diagnosis. COVID-19 diagnosis associated with .7269 (95% CI, 0.7088 to 0.7449 visits; P < .001) more total healthcare visits and an additional $223.60 (95% CI, 218.34 to 228.85; P < .001) in monthly costs. Excess monthly utilization and costs for individuals 17 years old and under subside after 5 months to .070 visits and $2.77, persist at substantial levels for all other groups and most pronounced among individuals age 45-64 (.207 visits and $73.43) and 65 years or older (.133 visits and $60.49). CONCLUSIONS This study found that COVID-19 diagnosis was associated with increased healthcare utilization and costs over a six-month post-diagnosis period. These findings imply a prolonged burden to the US healthcare system from medical encounters of COVID-19 patients and increased spending.
Collapse
Affiliation(s)
- Antonios M Koumpias
- Department of Social Sciences, University of Michigan-Dearborn, Dearborn, USA
| | - David Schwartzman
- Olin College of Business, Washington University in St. Louis, St. Louis, USA
| | - Owen Fleming
- Department of Economics, Wayne State University, 656 W. Kirby St FAB 2140, Detroit, USA.
| |
Collapse
|
22
|
Chiang KC, Kalantar-Zadeh K, Gupta A. Thymic Dysfunction and Atrophy in COVID-19 Disease Complicated by Inflammation, Malnutrition and Cachexia. Nutr Health 2022; 28:199-206. [PMID: 35234100 PMCID: PMC8891908 DOI: 10.1177/02601060221083160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Background: The current COVID-19 pandemic has put millions of people, especially children at risk of protein-energy malnutrition (PEM) by pushing them into poverty and disrupting the global food supply chain. The thymus is severely affected by nutritional deficiencies and is known as a barometer of malnutrition. Aim: The present commentary provides a novel perspective on the role of malnutrition-induced thymic dysfunction, involution and atrophy on the risk and severity of disease in children during the COVID-19 pandemic. Methods: A review of pertinent indexed literature including studies examining the effects of malnutrition on the thymus and immune dysfunction in COVID-19. Results: Protein-energy malnutrition and micronutrient deficiencies of zinc, iron and vitamin A are known to promote thymic dysfunction and thymocyte loss in children. Malnutrition- and infection-induced thymic atrophy and immune dysfunction may increase the risk of first, progression of COVID-19 disease to more severe forms including development of multisystem inflammatory syndrome in children (MIS-C); second, slow the recovery from COVID-19 disease; and third, increase the risk of other infections. Furthermore, malnourished children may be at increased risk of contracting SARS-CoV-2 infection due to socioeconomic conditions that promote viral transmission amongst contacts and create barriers to vaccination. Conclusion: National governments and international organizations including WHO, World Food Program, and UNICEF should institute measures to ensure provision of food and micronutrients for children at risk in order to limit the health impact of the ongoing COVID-19 pandemic.
Collapse
Affiliation(s)
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology, Hypertension
and Kidney Transplantation and Department of Medicine,
University of
California Irvine (UCI) School of Medicine,
USA
| | - Ajay Gupta
- Charak
Foundation, Orange, CA, USA
- Division of Nephrology, Hypertension
and Kidney Transplantation and Department of Medicine,
University of
California Irvine (UCI) School of Medicine,
USA
| |
Collapse
|
23
|
Post-COVID-19 Lymphocytopenia and Opportunistic Pathogens Infection in a Thalassemia Major Patient. THALASSEMIA REPORTS 2022. [DOI: 10.3390/thalassrep12020006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Transfusion-dependent thalassemia patients undergo transfusion immunomodulating effects, which result in a general immune response depression and, consequently, an increase in the frequency of infectious episodes and neoplastic events due to a reduction in phagocytic function. Altered natural killer functions and IL-2-mediated lymphocytic response, defects in antigen presentation due to monocyte–macrophage cells, and decreases in bone marrow precursors and HLA II+ cells all play key roles in immunodepression in thalassemia major. SARS-CoV-2 infection presents marked lymphopenia, occurring in 96.1% of severe cases. COVID-19-related lymphopenia is due to various mechanisms, which lead to an increase in lymphocytic apoptosis. Post-COVID-19 lymphocytic quantitative and functional disorders may compromise immune response and promote the onset of infections via opportunistic pathogens. Herein, we report a case of a thalassemia major patient who developed severe post-COVID-19 lymphocytopenia, which may have facilitated the onset of a severe Klebsiella Pneumoniae infection.
Collapse
|
24
|
Vollenberg R, Tepasse PR, Lorentzen E, Nowacki TM. Impaired Humoral Immunity with Concomitant Preserved T Cell Reactivity in IBD Patients on Treatment with Infliximab 6 Month after Vaccination with the SARS-CoV-2 mRNA Vaccine BNT162b2: A Pilot Study. J Pers Med 2022; 12:jpm12050694. [PMID: 35629116 PMCID: PMC9146879 DOI: 10.3390/jpm12050694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction: The Coronavirus Disease 2019 (COVID-19) pandemic has been caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The most important approach to prevent severe disease progression and to contain the pandemic is the use of COVID-19 vaccines. The aim of this study was to investigate the humoral and cellular response in immunosuppressed patients with inflammatory bowel disease (IBD) on treatment with anti-TNF (infliximab, adalimumab) and anti-α4ß7-Integrin (vedolizumab) 6 months after mRNA vaccination against SARS-CoV-2 compared to healthy subjects. Methods: In this prospective study, 20 IBD patients and 9 healthy controls were included 6 months after the second BNT162b2 vaccination. In addition to quantitative determination of IgG antibody levels against the SARS-CoV-2 receptor-binding domain (RBD) of the spike protein subunit S1, a SARS-CoV-2 surrogate neutralization test (sVNT) was used to assess potential neutralization capacity. SARS-CoV-2-specific T-cell responses were measured using an interferon-γ (IFN-γ) release assay (IGRA; Euroimmun Medical Laboratory Diagnostics, Lübeck, Germany). Results: S-IgG could still be detected in the majority of IBD patients 6 months after second vaccination. Compared to healthy controls, IBD patients treated with anti-TNF agents showed both lower neutralizing activity in sVNT (percent inhibition of ACE2 receptor binding by RBD protein) and lower IgG-S (AU/mL) antibody levels (AB) (sVNT: 79% vs. 2%, p < 0. 001; AB: 1018 AU/mL vs. 141 AU/mL, p = 0.025). In contrast, patients on therapy with vedolizumab showed no impairment in humoral immune response (sVNT, S-IgG) compared with healthy controls. Specific T-cellular reactivity was detected in 73% of IBD patients and in 67% of healthy controls independent of immunosuppressive therapy (anti-TNF., vedolizumab) (p = 0.189). Conclusion: Six months after BNT162b2 vaccination, this study found significantly decreased antibody levels in patients under anti-TNF therapy. IBD patients under anti-TNF and vedolizumab therapy had no impairment of T-cellular reactivity compared to healthy controls at this time point. Further studies with larger collectives for confirmation should follow.
Collapse
Affiliation(s)
- Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology, University Hospital Muenster, 48149 Muenster, Germany; (P.-R.T.); (T.M.N.)
- Correspondence:
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology, University Hospital Muenster, 48149 Muenster, Germany; (P.-R.T.); (T.M.N.)
| | - Eva Lorentzen
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Tobias Max Nowacki
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology, University Hospital Muenster, 48149 Muenster, Germany; (P.-R.T.); (T.M.N.)
- Department of Medicine, Gastroenterology, Marienhospital Steinfurt, 48565 Steinfurt, Germany
| |
Collapse
|
25
|
dos Santos PK, Sigoli E, Bragança LJ, Cornachione AS. The Musculoskeletal Involvement After Mild to Moderate COVID-19 Infection. Front Physiol 2022; 13:813924. [PMID: 35492595 PMCID: PMC9040683 DOI: 10.3389/fphys.2022.813924] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/01/2022] [Indexed: 12/13/2022] Open
Abstract
COVID-19, a disease caused by the novel coronavirus SARS-CoV-2, has been drastically affecting the daily lives of millions of people. COVID-19 is described as a multiorgan disease that affects not only the respiratory tract of infected individuals, but it has considerable effects on the musculoskeletal system, causing excessive fatigue, myalgia, arthralgia, muscle weakness and skeletal muscle damage. These symptoms can persist for months, decreasing the quality of life of numerous individuals. Curiously, most studies in the scientific literature focus on patients who were hospitalized due to SARS-CoV-2 infection and little is known about the mechanism of action of COVID-19 on skeletal muscles, especially of individuals who had the mild to moderate forms of the disease (non-hospitalized patients). In this review, we focus on the current knowledge about the musculoskeletal system in COVID-19, highlighting the lack of researches investigating the mild to moderate cases of infection and pointing out why it is essential to care for these patients. Also, we will comment about the need of more experimental data to assess the musculoskeletal manifestations on COVID-19-positive individuals.
Collapse
Affiliation(s)
- Patty K. dos Santos
- Muscle Physiology and Biophysics Laboratory, Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | | | | | - Anabelle S. Cornachione
- Muscle Physiology and Biophysics Laboratory, Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| |
Collapse
|
26
|
Kolesnyk M, Stepanova N. Post-COVID syndrome in dialysis patients and kidney transplant recipients. UKRAINIAN JOURNAL OF NEPHROLOGY AND DIALYSIS 2022:90-98. [DOI: 10.31450/ukrjnd.1(73).2022.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Abstract. Patients on maintenance dialysis treatment and kidney transplant recipients who survive coronavirus disease 2019 (COVID-19) are at higher risk of post-COVID syndrome compared to the general population. However, a detailed assessment of the causes, features, and clinical outcomes of the post-COVID syndrome in this patients’ cohort does not yet been established. In this review, we summarize published research on this issue to use these available data to predict the development, treatment and prevention of the post-COVID syndrome in dialysis patients and kidney transplant recipients.
Collapse
|
27
|
Acosta-Ampudia Y, Monsalve DM, Rojas M, Rodríguez Y, Zapata E, Ramírez-Santana C, Anaya JM. Persistent Autoimmune Activation and Proinflammatory State in Post-COVID Syndrome. J Infect Dis 2022; 225:2155-2162. [PMID: 35079804 PMCID: PMC8903340 DOI: 10.1093/infdis/jiac017] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
The immunopathological pathways enabling post-COVID syndrome (PCS) development are not entirely known. We underwent a longitudinal analysis of patients with COVID-19 who developed PCS aiming to evaluate the autoimmune and immunological status associated with this condition.
Methods
Thirty-three patients were included for longitudinal clinical and autoantibody analyses of whom 12 patients were assessed for cytokines and lymphocyte populations. Patients were followed during 7-11 months after acute COVID-19. Autoimmune profile and immunological status were evaluated mainly by enzyme-linked-immunosorbent assays and flow cytometry.
Results
Latent autoimmunity and overt autoimmunity persisted over time. A proinflammatory state was observed in patients with PCS characterized by upregulated IFN-α, TNF-α, G-CSF, IL-17A, IL-6, IL-1β, and IL-13, whereas IP-10 was decreased. In addition, PCS was characterized by increased levels of Th9, CD8+ effector T cells, naive B cells, and CD4+ effector memory T cells. Total levels of IgG S1-SARS-CoV-2 antibodies remained elevated over time.
Discussion
The clinical manifestations of PCS are associated with the persistence of a proinflammatory, and effector phenotype induced by SARS-CoV-2 infection. This long-term persistent immune activation may contribute to the development of latent and overt autoimmunity. Results suggest the need to evaluate the role of immunomodulation in the treatment of PCS.
Collapse
Affiliation(s)
- Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
- Clínica del Occidente, Bogota, Colombia
| | - Elizabeth Zapata
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
- Clínica del Occidente, Bogota, Colombia
| |
Collapse
|
28
|
Jones OY, Yeralan S. Is Long COVID a State of Systemic Pericyte Disarray? J Clin Med 2022; 11:jcm11030572. [PMID: 35160024 PMCID: PMC8836446 DOI: 10.3390/jcm11030572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 01/03/2023] Open
Abstract
The most challenging aspect of Post-Acute Sequelae of SARS-CoV-2 Infection (PASC) or Long COVID remains for the discordance between the viral damage from acute infection in the recent past and susceptibility of Long COVID without clear evidence of post infectious inflammation or autoimmune reactions. In this communication we propose that disarray of pericytes plays a central role in emerge of Long COVID. We assume pericytes are agents with “Triple-A” qualities, i.e., analyze-adapt and advance, necessary for sustainability of host homeostasis. Based on this view, we further suggest Long COVID may provide a model system to integrate system theory and complex adaptive systems to explore a new class of maladies those are currently not well defined and with no remedies.
Collapse
Affiliation(s)
- Olcay Y. Jones
- Pediatric Rheumatology, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Correspondence: (O.Y.J.); (S.Y.)
| | - Sencer Yeralan
- School of IT and Engineering, ADA University, Baku AZ1008, Azerbaijan
- Correspondence: (O.Y.J.); (S.Y.)
| |
Collapse
|
29
|
Vollenberg R, Tepasse PR, Kühn JE, Hennies M, Strauss M, Rennebaum F, Schomacher T, Boeckel G, Lorentzen E, Bokemeyer A, Nowacki TM. Humoral Immune Response in IBD Patients Three and Six Months after Vaccination with the SARS-CoV-2 mRNA Vaccines mRNA-1273 and BNT162b2. Biomedicines 2022; 10:171. [PMID: 35052849 PMCID: PMC8774019 DOI: 10.3390/biomedicines10010171] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome coronovirus-2 (SARS-CoV-2) is the cause of the coronavirus disease 2019 (COVID-19) pandemic. Vaccination is considered the core approach to containing the pandemic. There is currently insufficient evidence on the efficacy of these vaccines in immunosuppressed inflammatory bowel disease (IBD) patients. The aim of this study was to investigate the humoral response in immunosuppressed IBD patients after COVID-19 mRNA vaccination. In this prospective study, IgG antibody levels (AB) against the SARS-CoV-2 receptor-binding domain (spike-protein) were quantitatively determined. For assessing the potential neutralizing capacity, a SARS-CoV-2 surrogate neutralization test (sVNT) was employed in IBD patients (n = 95) and healthy controls (n = 38). Sera were examined prior to the first/second vaccination and 3/6 months after second vaccination. Patients showed lower sVNT (%) and IgG-S (AU/mL) AB both before the second vaccination (sVNT p < 0.001; AB p < 0.001) and 3 (sVNT p = 0.002; AB p = 0.001) and 6 months (sVNT p = 0.062; AB p = 0.061) after the second vaccination. Although seroconversion rates (sVNT, IgG-S) did not differ between the two groups 3 months after second vaccination, a significant difference was seen 6 months after second vaccination (sVNT p = 0.045). Before and three months after the second vaccination, patients treated with anti-tumor necrosis factor (TNF) agents showed significantly lower AB than healthy subjects. In conclusion, an early booster shot vaccination should be discussed for IBD patients on anti-TNF therapy.
Collapse
Affiliation(s)
- Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Joachim Ewald Kühn
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany; (J.E.K.); (M.H.); (E.L.)
| | - Marc Hennies
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany; (J.E.K.); (M.H.); (E.L.)
| | - Markus Strauss
- Department of Medicine C, Cardiology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Florian Rennebaum
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Tina Schomacher
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
| | - Göran Boeckel
- Department of Medicine D, Division of General Internal and Emergency Medicine, Nephrology and Rheumatology, University Hospital Muenster, 48149 Muenster, Germany;
| | - Eva Lorentzen
- Institute of Virology, University Hospital Muenster, 48149 Muenster, Germany; (J.E.K.); (M.H.); (E.L.)
| | - Arne Bokemeyer
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, 45147 Essen, Germany;
| | - Tobias Max Nowacki
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clincial Infectiology University Hospital Muenster, 48149 Muenster, Germany; (F.R.); (T.S.); (T.M.N.)
- Department of Medicine, Gastroenterology, Marienhospital Steinfurt, 48565 Steinfurt, Germany
| |
Collapse
|
30
|
Morbelli S, Chiola S, Donegani MI, Arnaldi D, Pardini M, Mancini R, Lanfranchi F, D'amico F, Bauckneht M, Miceli A, Biassoni E, Orso B, Barisione E, Benedetti L, Gianmario S, Nobili F. Metabolic correlates of olfactory dysfunction in COVID-19 and Parkinson's disease (PD) do not overlap. Eur J Nucl Med Mol Imaging 2022; 49:1939-1950. [PMID: 34984501 PMCID: PMC8727173 DOI: 10.1007/s00259-021-05666-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/19/2021] [Indexed: 12/20/2022]
Abstract
Purpose Hyposmia is a common feature of COVID-19 and Parkinson’s disease (PD). As parkinsonism has been reported after COVID-19, a link has been hypothesized between SARS-CoV2 infection and PD. We aimed to evaluate brain metabolic correlates of isolated persistent hyposmia after mild-to-moderate COVID-19 and to compare them with metabolic signature of hyposmia in drug-naïve PD patients. Methods Forty-four patients who experienced hyposmia after SARS-COV2 infection underwent brain [18F]-FDG PET in the first 6 months after recovery. Olfaction was assessed by means of the 16-item “Sniffin’ Sticks” test and patients were classified as with or without persistent hyposmia (COVID-hyposmia and COVID-no-hyposmia respectively). Brain [18F]-FDG PET of post-COVID subgroups were compared in SPM12. COVID-hyposmia patients were also compared with eighty-two drug-naïve PD patients with hyposmia. Multiple regression analysis was used to identify correlations between olfactory test scores and brain metabolism in patients’ subgroups. Results COVID-hyposmia patients (n = 21) exhibited significant hypometabolism in the bilateral gyrus rectus and orbitofrontal cortex with respect to COVID-non-hyposmia (n = 23) (p < 0.002) and in middle and superior temporal gyri, medial/middle frontal gyri, and right insula with respect to PD-hyposmia (p < 0.012). With respect to COVID-hyposmia, PD-hyposmia patients showed hypometabolism in inferior/middle occipital gyri and cuneus bilaterally. Olfactory test scores were directly correlated with metabolism in bilateral rectus and medial frontal gyri and in the right middle temporal and anterior cingulate gyri in COVID-hyposmia patients (p < 0.006) and with bilateral cuneus/precuneus and left lateral occipital cortex in PD-hyposmia patients (p < 0.004). Conclusion Metabolic signature of persistent hyposmia after COVID-19 encompasses cortical regions involved in olfactory perception and does not overlap metabolic correlates of hyposmia in PD.
Collapse
Affiliation(s)
- Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Genova, Italy. .,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy.
| | - Silvia Chiola
- Humanitas Clinical and Research Center-IRCCS, Via Manzoni 56, Rozzano, 20089, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Italy
| | - Maria Isabella Donegani
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Dario Arnaldi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, 516126, Genoa, Italy
| | - Matteo Pardini
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, 516126, Genoa, Italy
| | - Raffaele Mancini
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, 516126, Genoa, Italy
| | - Francesco Lanfranchi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Francesca D'amico
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Alberto Miceli
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Erica Biassoni
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, 516126, Genoa, Italy
| | - Beatrice Orso
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, 516126, Genoa, Italy
| | - Emanuela Barisione
- Interventional Pulmonology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luana Benedetti
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, 516126, Genoa, Italy
| | - Sambuceti Gianmario
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Flavio Nobili
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, 516126, Genoa, Italy
| |
Collapse
|
31
|
Luchian ML, Motoc A, Lochy S, Magne J, Belsack D, De Mey J, Roosens B, Van den Bussche K, Boeckstaens S, Chameleva H, Geers J, Houard L, De Potter T, Allard S, Weytjens C, Droogmans S, Cosyns B. Subclinical Myocardial Dysfunction in Patients with Persistent Dyspnea One Year after COVID-19. Diagnostics (Basel) 2021; 12:diagnostics12010057. [PMID: 35054224 PMCID: PMC8775030 DOI: 10.3390/diagnostics12010057] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/16/2022] Open
Abstract
Long coronavirus disease 2019 (COVID-19) was described in patients recovering from COVID-19, with dyspnea being a frequent symptom. Data regarding the potential mechanisms of long COVID remain scarce. We investigated the presence of subclinical cardiac dysfunction, assessed by transthoracic echocardiography (TTE), in recovered COVID-19 patients with or without dyspnea, after exclusion of previous cardiopulmonary diseases. A total of 310 consecutive COVID-19 patients were prospectively included. Of those, 66 patients (mean age 51.3 ± 11.1 years, almost 60% males) without known cardiopulmonary diseases underwent one-year follow-up consisting of clinical evaluation, spirometry, chest computed tomography, and TTE. From there, 23 (34.8%) patients reported dyspnea. Left ventricle (LV) ejection fraction was not significantly different between patients with or without dyspnea (55.7 ± 4.6 versus (vs.) 57.6 ± 4.5, p = 0.131). Patients with dyspnea presented lower LV global longitudinal strain, global constructive work (GCW), and global work index (GWI) compared to asymptomatic patients (−19.9 ± 2.1 vs. −21.3 ± 2.3 p = 0.039; 2183.7 ± 487.9 vs. 2483.1 ± 422.4, p = 0.024; 1960.0 ± 396.2 vs. 2221.1 ± 407.9, p = 0.030). GCW and GWI were inversely and independently associated with dyspnea (p = 0.035, OR 0.998, 95% CI 0.997–1.000; p = 0.040, OR 0.998, 95% CI 0.997–1.000). Persistent dyspnea one-year after COVID-19 was present in more than a third of the recovered patients. GCW and GWI were the only echocardiographic parameters independently associated with symptoms, suggesting a decrease in myocardial performance and subclinical cardiac dysfunction.
Collapse
Affiliation(s)
- Maria-Luiza Luchian
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
- Correspondence:
| | - Andreea Motoc
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Stijn Lochy
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Julien Magne
- Service Cardiologie, CHU Limoges, Hôpital Dupuytren, 87000 Limoges, France;
- 16 INSERM 1094 Faculté de Médecine de Limoges, 87000 Limoges, France
| | - Dries Belsack
- Department of Radiology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (D.B.); (J.D.M.)
| | - Johan De Mey
- Department of Radiology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (D.B.); (J.D.M.)
| | - Bram Roosens
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Karen Van den Bussche
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Sven Boeckstaens
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Hadischat Chameleva
- Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (H.C.); (T.D.P.)
| | - Jolien Geers
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Laura Houard
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Tom De Potter
- Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (H.C.); (T.D.P.)
| | - Sabine Allard
- Department of Internal Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium;
| | - Caroline Weytjens
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Steven Droogmans
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| | - Bernard Cosyns
- Department of Cardiology, (Centrum voor Hart-en Vaatziekten), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (A.M.); (S.L.); (B.R.); (K.V.d.B.); (S.B.); (J.G.); (L.H.); (C.W.); (S.D.); (B.C.)
| |
Collapse
|
32
|
Aguillón-Durán GP, Prieto-Martínez E, Ayala D, García J, Thomas JM, García JI, Henry BM, Torrelles JB, Turner J, Ledezma-Campos E, Restrepo BI. COVID-19 and chronic diabetes: the perfect storm for reactivation tuberculosis?: a case series. J Med Case Rep 2021; 15:621. [PMID: 34915933 PMCID: PMC8674832 DOI: 10.1186/s13256-021-03193-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/16/2021] [Indexed: 11/24/2022] Open
Abstract
Background The coronavirus disease 2019 pandemic is predicted to have a net negative effect on tuberculosis control, with an estimated excess of 6.3 million tuberculosis cases and 1.4 million deaths by 2025. Programmatic issues such as the lockdown of tuberculosis services affect all patients, while biosocial factors have a differential impact on an individual’s risk for tuberculosis or adverse tuberculosis outcomes. Case presentation We report three Hispanic cases of incident tuberculosis (two males, 43 and 44 years old; one female, 49 years old) after resolution of coronavirus disease episodes. Coincidentally, all cases shared a common risk factor: a chronic history poorly controlled diabetes. Conclusions Our findings alert to the threat posed by the synergy between coronavirus disease and diabetes, on tuberculosis reactivation. In medium- to high-risk settings for tuberculosis, we recommend implementation of routine screening for latent tuberculosis infection in these cases, and preventive tuberculosis treatment in those who are positive.
Collapse
Affiliation(s)
- Genesis P Aguillón-Durán
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX, 78520, USA
| | - Ericka Prieto-Martínez
- Secretaria de Salud de Tamaulipas, 88630, Reynosa, Tamaulipas, Mexico.,Secretaria de Salud de Tamaulipas, 87000, Ciudad Victoria, Tamaulipas, Mexico
| | - Doris Ayala
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX, 78520, USA
| | - Juan García
- Department of Human Genetics, South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, 1214 W Schunior, UTRGV-Edinburg Campus, Edinburg, TX, 78541, USA
| | - John M Thomas
- Department of Human Genetics, South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, 1214 W Schunior, UTRGV-Edinburg Campus, Edinburg, TX, 78541, USA
| | - Juan Ignacio García
- Population Health Program and Host Pathogens Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, 78229, USA
| | - Brandon Michael Henry
- Population Health Program and Host Pathogens Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, 78229, USA
| | - Jordi B Torrelles
- Population Health Program and Host Pathogens Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, 78229, USA
| | - Joanne Turner
- Population Health Program and Host Pathogens Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, 78229, USA
| | - Eder Ledezma-Campos
- Department of Human Genetics, South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, 1214 W Schunior, UTRGV-Edinburg Campus, Edinburg, TX, 78541, USA
| | - Blanca I Restrepo
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Campus, Brownsville, TX, 78520, USA. .,Department of Human Genetics, South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, 1214 W Schunior, UTRGV-Edinburg Campus, Edinburg, TX, 78541, USA.
| |
Collapse
|