451
|
Costantino M, Giudice V, Farroni M, Marongiu F, De Caro F, Filippelli A. Impact of Spa Therapy on Symptoms and Quality of Life in Post-COVID-19 Patients with Chronic Conditions. J Clin Med 2024; 13:5091. [PMID: 39274303 PMCID: PMC11396595 DOI: 10.3390/jcm13175091] [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: 06/27/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/16/2024] Open
Abstract
Background: With limited pharmacological interventions, post-COVID-19 condition is a clinical challenge, and supplementary therapies are essential for symptom relief and enhancing quality of life (QoL). In our prospective observational study, we aimed to evaluate the impact of Salus per aquam (Spa) therapy on post-COVID-19 symptoms and QoL in individuals who suffer from chronic joint, musculoskeletal, skin, and/or respiratory conditions. Methods: A total of 159 individuals undergoing Spa therapy were enrolled, and 78 of them had post-COVID-19 symptoms, assessed using Visual Analogue Scale (VAS) and modified British Medical Research Council Questionnaire (mMRC-DS scales), as well as the Short Form 36 Health Status Survey (SF-36) questionnaire for QoL. Results: Spa therapy significantly reduced most post-COVID-19 symptoms, especially chronic fatigue, pain, brain fog, and persistent cough (all p < 0.05), as well as physical (+72%) and emotional (+66%) limitations. When stratified by sex, males showed a greater improvement from baseline, while females consistently displayed a higher amelioration in all QoL dimensions. Moreover, full vaccination with 3-4 doses significantly protected against SARS-CoV-2 re-infections and post-COVID-19 development (p < 0.05). Conclusions: Spa therapy demonstrated effectiveness in mitigating post-COVID-19 symptoms and enhancing QoL in patients suffering from chronic diseases.
Collapse
Affiliation(s)
- Maria Costantino
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy
- University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84121 Salerno, Italy
- Non-Profit Association F.I.R.S.Thermae (Interdisciplinary Training, Researches and Spa Sciences), 80078 Pozzuoli, Italy
| | - Valentina Giudice
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy
- University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84121 Salerno, Italy
| | - Mario Farroni
- Non-Profit Association F.I.R.S.Thermae (Interdisciplinary Training, Researches and Spa Sciences), 80078 Pozzuoli, Italy
| | - Francesco Marongiu
- Non-Profit Association F.I.R.S.Thermae (Interdisciplinary Training, Researches and Spa Sciences), 80078 Pozzuoli, Italy
| | - Francesco De Caro
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy
- University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84121 Salerno, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84081 Baronissi, Italy
- University Hospital "San Giovanni di Dio e Ruggi d'Aragona", 84121 Salerno, Italy
| |
Collapse
|
452
|
Frasson I, Diamante L, Zangrossi M, Carbognin E, Pietà AD, Penna A, Rosato A, Verin R, Torrigiani F, Salata C, Dizanzo MP, Vaccaro L, Cacchiarelli D, Richter SN, Montagner M, Martello G. Identification of druggable host dependency factors shared by multiple SARS-CoV-2 variants of concern. J Mol Cell Biol 2024; 16:mjae004. [PMID: 38305139 PMCID: PMC11411213 DOI: 10.1093/jmcb/mjae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/23/2023] [Accepted: 01/31/2024] [Indexed: 02/03/2024] Open
Abstract
The high mutation rate of SARS-CoV-2 leads to the emergence of multiple variants, some of which are resistant to vaccines and drugs targeting viral elements. Targeting host dependency factors, e.g. cellular proteins required for viral replication, would help prevent the development of resistance. However, it remains unclear whether different SARS-CoV-2 variants induce conserved cellular responses and exploit the same core host factors. To this end, we compared three variants of concern and found that the host transcriptional response was conserved, differing only in kinetics and magnitude. Clustered regularly interspaced short palindromic repeats screening identified host genes required for each variant during infection. Most of the genes were shared by multiple variants. We validated our hits with small molecules and repurposed the US Food and Drug Administration-approved drugs. All the drugs were highly active against all the tested variants, including new variants that emerged during the study (Delta and Omicron). Mechanistically, we identified reactive oxygen species production as a key step in early viral replication. Antioxidants such as N-acetyl cysteine (NAC) were effective against all the variants in both human lung cells and a humanized mouse model. Our study supports the use of available antioxidant drugs, such as NAC, as a general and effective anti-COVID-19 approach.
Collapse
Affiliation(s)
- Ilaria Frasson
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Linda Diamante
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Manuela Zangrossi
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Elena Carbognin
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Alessandro Penna
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua 35128, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | | | - Lorenzo Vaccaro
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
- School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples Federico II, Naples 80138, Italy
| | - Sara N Richter
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Microbiology and Virology Unit, Padua University Hospital, Padua 35128, Italy
| | - Marco Montagner
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Graziano Martello
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| |
Collapse
|
453
|
Hwang JH, Lee KN, Kim SM, Kim H, Park SH, Kim DW, Cho G, Lee YH, Lee JS, Park JH. Enhanced Effects of ISA 207 Adjuvant via Intradermal Route in Foot-and-Mouth Disease Vaccine for Pigs. Vaccines (Basel) 2024; 12:963. [PMID: 39339996 PMCID: PMC11435775 DOI: 10.3390/vaccines12090963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
In South Korea, a mandatory nation-wide foot-and-mouth disease (FMD) vaccination policy is in place. However, a major side effect of the current method of intramuscular (IM) administration of oil-adjuvanted FMD vaccines is the formation of granulomas in the muscles of pigs. To address this issue, we assessed the possible application of intradermal (ID) vaccination. Initially, we compared the serological immune response in specific pathogen-free pigs inoculated with FMD vaccines formulated with eight different adjuvants, administered twice at the neck site using a syringe with a needle via the ID route. Among the formulations (water-in-oil-in-water (W/O/W), oil-in-water (O/W), and polymer nanomaterials), ISA 207 of W/O/W was the most effective in inducing immunogenicity followed by ISA 201 of W/O/W. ISA 207 was further tested in formulations of different antigen doses (12 or 1.2 μg) delivered via both IM and ID routes. All four treatments successfully protected the pigs against FMD virus challenges. To assess the feasibility of the field application of the vaccines with ISA 207, we conducted ID vaccination of conventional pigs using a needle-free device, resulting in the detection of significant levels of neutralizing antibodies. ISA 207 was shown to be superior to ISA 201 in inducing immunogenicity via the ID route. In conclusion, ISA 207 could be a suitable adjuvant for ID vaccination in terms of vaccine efficacy for FMD, allowing for alternate use of ID vaccination and subsequent reduction in the incidences of granuloma formation in the field.
Collapse
Affiliation(s)
- Ji-Hyeon Hwang
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
- Veterinary College, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kwang-Nyeong Lee
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| | - Su-Mi Kim
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| | - Hyejin Kim
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| | - Sung-Han Park
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| | - Dong-Wan Kim
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| | - Giyoun Cho
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| | - Yoon-Hee Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| | - Jong-Soo Lee
- Veterinary College, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jong-Hyeon Park
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, 177 Hyeoksin 8, Gimcheon 39660, Republic of Korea
| |
Collapse
|
454
|
Zhai X, Wu W, Zeng S, Miao Y. Advance in the mechanism and clinical research of myalgia in long COVID. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:142-164. [PMID: 39310121 PMCID: PMC11411160 DOI: 10.62347/txvo6284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 07/18/2024] [Indexed: 09/25/2024]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve, mortality rates of coronavirus disease 2019 (COVID-19) have significantly decreased. However, a variable proportion of patients exhibit persistent prolonged symptoms of COVID-19 infection (long COVID). This virus primarily attacks respiratory system, but numerous individuals complain persistent skeletal muscle pain or worsening pre-existing muscle pain post COVID-19, which severely affects the quality of life and recovery. Currently, there is limited research on the skeletal muscle pain in long COVID. In this brief review, we review potential pathological mechanisms of skeletal muscle pain in long COVID, and summarize the various auxiliary examinations and treatments for skeletal muscle pain in long COVID. We consider abnormal activation of inflammatory response, myopathy, and neurological damages as pivotal pathological mechanisms of skeletal muscle pain in long COVID. A comprehensive examination is significantly important in order to work out effective treatment plans and relieve skeletal muscle pain. So far, rehabilitation interventions for myalgia in long COVID contain but are not limited to drug, nutraceutical therapy, gut microbiome-targeted therapy, interventional therapy and strength training. Our study provides a potential mechanism reference for clinical researches, highlighting the importance of comprehensive approach and management of skeletal muscle pain in long COVID. The relief of skeletal muscle pain will accelerate rehabilitation process, improve activities of daily living and enhance the quality of life, promoting individuals return to society with profound significance.
Collapse
Affiliation(s)
- Xiuyun Zhai
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
| | - Weijun Wu
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
| | - Siliang Zeng
- Department of Rehabilitation Therapy, School of Health, Shanghai Normal University Tianhua CollegeNo. 1661, North Shengxin Road, Shanghai 201815, China
| | - Yun Miao
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
- Department of Rehabilitation, School of International Medical Technology, Shanghai Sanda UniversityNo. 2727, Jinhai Road, Shanghai 201209, China
| |
Collapse
|
455
|
Wang H, Hu J, Zhou W, Qian A. Metabolic reprogramming in the pathogenesis and progression of nasopharyngeal carcinoma: molecular mechanisms and therapeutic implications. Am J Cancer Res 2024; 14:4049-4064. [PMID: 39267663 PMCID: PMC11387871 DOI: 10.62347/vyat9271] [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: 06/14/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a unique head and neck cancer with a complex etiology involving genetic predispositions, environmental factors, and Epstein-Barr virus (EBV) infection. Despite progress in radiotherapy and chemotherapy, the prognosis for advanced NPC is still unfavorable, prompting the need for innovative therapeutic approaches. Metabolic reprogramming plays a crucial role in the development and progression of NPC, marked by substantial changes in glycolysis, lipid, and amino acid metabolism. These alterations aid tumor cell proliferation, survival under stress, and immune evasion, with features such as enhanced aerobic glycolysis (Warburg effect) and shifts in lipid and amino acid pathways. Oncogenic drivers like MYC, RAS, EGFR, and the loss of tumor suppressors such as TP53 and PTEN, along with key signaling pathways including mTOR, AMPK, and HIF-1α, orchestrate these metabolic changes. This review discusses the molecular mechanisms of metabolic reprogramming in NPC and outlines potential therapeutic targets within these pathways. Advances in metabolic imaging and biomarker discovery are also enhancing the precision of diagnostics and treatment monitoring, fostering personalized medicine in NPC treatment. This manuscript aims to provide a detailed overview of the current research and its implications for improving NPC management and patient outcomes through targeted metabolic therapies.
Collapse
Affiliation(s)
- Hongli Wang
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Jiandao Hu
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Weibang Zhou
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Aijuan Qian
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| |
Collapse
|
456
|
Cai X, Lin W, Wu F, Song G, Qian Z, Wang Y. RARB associated with MSI, affects progression and prognosis of gastric cancer. BMC Gastroenterol 2024; 24:285. [PMID: 39179979 PMCID: PMC11342619 DOI: 10.1186/s12876-024-03339-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 07/25/2024] [Indexed: 08/26/2024] Open
Abstract
Microsatellite instability (MSI) has been widely acknowledged as an important factor regulating tumor intrinsic biological behavior and affecting the survival of gastric cancer patients. Here, we firstly identified the RARB as a gene associated with MSI gastric cancer. RARB was downregulated in human gastric cancer tissues compared to paired paracancerous tissues, Knockdown of RARB accelerated the proliferation, invasion and migration of cancer cells in vitro. Mechanismly, RARB knockdown promoted epithelial-mesenchymal transition (EMT) process of gastric cancer. However, RARBLow patients exhibited better survival compared to RARBHigh patients. Further study revealed that RARB expression was inversely correlated with MSI status and immune infiltrates in vivo. Thus, RARB may be a potential target for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Xufan Cai
- Graduate School, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Wenfa Lin
- Graduate School, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University Hangzhou, Hangzhou, Zhejiang, China
| | - Fang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, The Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Guangyuan Song
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery, Zhejiang Provincial People's Hospital, The Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhenyuan Qian
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, The Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yu Wang
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital, The Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
457
|
Letafati A, Taghiabadi Z, Roushanzamir M, Memarpour B, Seyedi S, Farahani AV, Norouzi M, Karamian S, Zebardast A, Mehrabinia M, Ardekani OS, Fallah T, Khazry F, Daneshvar SF, Norouzi M. From discovery to treatment: tracing the path of hepatitis E virus. Virol J 2024; 21:194. [PMID: 39180020 PMCID: PMC11342613 DOI: 10.1186/s12985-024-02470-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
The hepatitis E virus (HEV) is a major cause of acute viral hepatitis worldwide. HEV is classified into eight genotypes, labeled HEV-1 through HEV-8. Genotypes 1 and 2 exclusively infect humans, while genotypes 3, 4, and 7 can infect both humans and animals. In contrast, genotypes 5, 6, and 8 are restricted to infecting animals. While most individuals with a strong immune system experience a self-limiting infection, those who are immunosuppressed may develop chronic hepatitis. Pregnant women are particularly vulnerable to severe illness and mortality due to HEV infection. In addition to liver-related complications, HEV can also cause extrahepatic manifestations, including neurological disorders. The immune response is vital in determining the outcome of HEV infection. Deficiencies in T cells, NK cells, and antibody responses are linked to poor prognosis. Interestingly, HEV itself contains microRNAs that regulate its replication and modify the host's antiviral response. Diagnosis of HEV infection involves the detection of HEV RNA and anti-HEV IgM/IgG antibodies. Supportive care is the mainstay of treatment for acute infection, while chronic HEV infection may be cleared with the use of ribavirin and pegylated interferon. Prevention remains the best approach against HEV, focusing on sanitation infrastructure improvements and vaccination, with one vaccine already licensed in China. This comprehensive review provides insights into the spread, genotypes, prevalence, and clinical effects of HEV. Furthermore, it emphasizes the need for further research and attention to HEV, particularly in cases of acute hepatitis, especially among solid-organ transplant recipients.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran.
| | - Zahra Taghiabadi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mahshid Roushanzamir
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Department of Pharmacological and Biomolecular Science, University of Milan, Milan, Italy
| | - Bahar Memarpour
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Saba Seyedi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | | | - Masoomeh Norouzi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Saeideh Karamian
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Arghavan Zebardast
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Marzieh Mehrabinia
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Tina Fallah
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Fatemeh Khazry
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Samin Fathi Daneshvar
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mehdi Norouzi
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
458
|
Sato-Espinoza K, Berrospi D, Diaz-Ferrer J. Case report: liver failure as a debut of autoimmune hepatitis triggered by dengue virus in a pregnant woman. Rev Peru Med Exp Salud Publica 2024; 41:209-213. [PMID: 39166644 PMCID: PMC11300697 DOI: 10.17843/rpmesp.2024.412.13485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/17/2024] [Indexed: 08/23/2024] Open
Abstract
Autoimmune hepatitis (AIH) is a complex condition with unclear origins, involving genetic susceptibility and environmental triggers that lead to immune system dysfunction. We report a case of a pregnant woman from a mosquito-borne disease-endemic area who presented jaundice, abdominal pain, and pruritus, complicated by acute liver failure. Immunological markers showed AIH triggered by dengue virus infection, which was confirmed by a positive IgM test. Treatment with supportive care followed by steroids and azathioprine resulted in favorable outcomes, averting the need for a liver transplant. Although AIH can be triggered by viruses, the role of dengue in its pathogenesis remains poorly understood. Regular clinical monitoring is vital for managing AIH, particularly during pregnancy, due to variable immune status and treatment responses. Further research is necessary to understand the link between dengue infection and AIH. Individualized treatment strategies are crucial, especially during pregnancy, in order to ensure favorable outcomes.
Collapse
Affiliation(s)
- Karina Sato-Espinoza
- Escuela de Medicina. Universidad Peruana de Ciencias Aplicadas (UPC), Lima, Peru.Universidad Peruana de Ciencias AplicadasEscuela de MedicinaUniversidad Peruana de Ciencias Aplicadas (UPC)LimaPeru
| | - Diego Berrospi
- Facultad de Medicina, Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru.Universidad Peruana Cayetano HerediaFacultad de MedicinaUniversidad Peruana Cayetano Heredia (UPCH)LimaPeru
| | - Javier Diaz-Ferrer
- Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru.Hospital Nacional Edgardo Rebagliati MartinsLimaPeru
- Facultad de Medicina, Universidad San Martín de Porres, Lima, Peru.Universidad de San Martín de PorresFacultad de MedicinaUniversidad San Martín de PorresLimaPeru
| |
Collapse
|
459
|
Biswas M, Nurunnabi M, Khatun Z. Understanding Mucosal Physiology and Rationale of Formulation Design for Improved Mucosal Immunity. ACS APPLIED BIO MATERIALS 2024; 7:5037-5056. [PMID: 38787767 DOI: 10.1021/acsabm.4c00395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
The oral and nasal cavities serve as critical gateways for infectious pathogens, with microorganisms primarily gaining entry through these routes. Our first line of defense against these invaders is the mucosal membrane, a protective barrier that shields the body's internal systems from infection while also contributing to vital functions like air and nutrient intake. One of the key features of this mucosal barrier is its ability to protect the physiological system from pathogens. Additionally, mucosal tolerance plays a crucial role in maintaining homeostasis by regulating the pH and water balance within the body. Recognizing the importance of the mucosal barrier, researchers have developed various mucosal formulations to enhance the immune response. Mucosal vaccines, for example, deliver antigens directly to mucosal tissues, triggering local immune stimulation and ultimately inducing systemic immunity. Studies have shown that lipid-based formulations such as liposomes and virosomes can effectively elicit both local and systemic immune responses. Furthermore, mucoadhesive polymeric particles, with their prolonged delivery to target sites, have demonstrated an enhanced immune response. This Review delves into the critical role of material selection and delivery approaches in optimizing mucosal immunity.
Collapse
Affiliation(s)
- Mila Biswas
- Department of Electrical and Computer Engineering, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Zehedina Khatun
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| |
Collapse
|
460
|
Zhang M, Chen H, Liu H, Tang H. The impact of integrated hepatitis B virus DNA on oncogenesis and antiviral therapy. Biomark Res 2024; 12:84. [PMID: 39148134 PMCID: PMC11328401 DOI: 10.1186/s40364-024-00611-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/29/2024] [Indexed: 08/17/2024] Open
Abstract
The global burden of hepatitis B virus (HBV) infection remains high, with chronic hepatitis B (CHB) patients facing a significantly increased risk of developing cirrhosis and hepatocellular carcinoma (HCC). The ultimate objective of antiviral therapy is to achieve a sterilizing cure for HBV. This necessitates the elimination of intrahepatic covalently closed circular DNA (cccDNA) and the complete eradication of integrated HBV DNA. This review aims to summarize the oncogenetic role of HBV integration and the significance of clearing HBV integration in sterilizing cure. It specifically focuses on the molecular mechanisms through which HBV integration leads to HCC, including modulation of the expression of proto-oncogenes and tumor suppressor genes, induction of chromosomal instability, and expression of truncated mutant HBV proteins. The review also highlights the impact of antiviral therapy in reducing HBV integration and preventing HBV-related HCC. Additionally, the review offers insights into future objectives for the treatment of CHB. Current strategies for HBV DNA integration inhibition and elimination include mainly antiviral therapies, RNA interference and gene editing technologies. Overall, HBV integration deserves further investigation and can potentially serve as a biomarker for CHB and HBV-related HCC.
Collapse
Affiliation(s)
- Mingming Zhang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Han Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Huan Liu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China.
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
461
|
Arshad NF, Nordin FJ, Foong LC, In LLA, Teo MYM. Engineering receptor-binding domain and heptad repeat domains towards the development of multi-epitopes oral vaccines against SARS-CoV-2 variants. PLoS One 2024; 19:e0306111. [PMID: 39146295 PMCID: PMC11326571 DOI: 10.1371/journal.pone.0306111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 06/11/2024] [Indexed: 08/17/2024] Open
Abstract
The inability of existing vaccines to cope with the mutation rate has highlighted the need for effective preventative strategies for COVID-19. Through the secretion of immunoglobulin A, mucosal delivery of vaccines can effectively stimulate mucosal immunity for better protection against SARS-CoV-2 infection. In this study, various immunoinformatic tools were used to design a multi-epitope oral vaccine against SARS-CoV-2 based on its receptor-binding domain (RBD) and heptad repeat (HR) domains. T and B lymphocyte epitopes were initially predicted from the RBD and HR domains of SARS-CoV-2, and potential antigenic, immunogenic, non-allergenic, and non-toxic epitopes were identified. Epitopes that are highly conserved and have no significant similarity to human proteome were selected. The epitopes were joined with appropriate linkers, and an adjuvant was added to enhance the vaccine efficacy. The vaccine 3D structure constructs were docked with toll-like receptor 4 (TLR-4) and TLR1-TLR2, and the binding affinity was calculated. The designed multi-epitope vaccine construct (MEVC) consisted of 33 antigenic T and B lymphocyte epitopes. The results of molecular dockings and free binding energies confirmed that the MEVC effectively binds to TLR molecules, and the complexes were stable. The results suggested that the designed MEVC is a potentially safe and effective oral vaccine against SARS-CoV-2. This in silico study presents a novel approach for creating an oral multi-epitope vaccine against the rapidly evolving SARS-CoV-2 variants. These findings offer valuable insights for developing an effective strategy to combat COVID-19. Further preclinical and clinical studies are required to confirm the efficacy of the MEVC vaccine.
Collapse
Affiliation(s)
- Nur Farhanah Arshad
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Fariza Juliana Nordin
- Department of Biological Sciences and Biotechnology, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Lian Chee Foong
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Michelle Yee Mun Teo
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
462
|
Zima K, Khaidakov B, Sochocka M, Ochnik M, Lemke K, Kowalczyk P. Exploring the potency of polyphenol-rich blend from Lonicera caerulea var. Kamtschatica sevast., Aronia melanocarpa, and Echinacea purpurea: Promising anti-inflammatory, antioxidant, and antiviral properties. Heliyon 2024; 10:e35630. [PMID: 39170264 PMCID: PMC11336823 DOI: 10.1016/j.heliyon.2024.e35630] [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: 03/25/2024] [Revised: 06/25/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Previous studies have highlighted the beneficial properties of plants rich in polyphenols, such as Lonicera caerulea var. Kamtschatica Sevast. (LCK), Aronia melanocarpa (AM), and Echinacea purpurea (EP). These plants have demonstrated antioxidant, immunomodulatory, and potential antiviral effects. Thus, the objective of this study was to investigate the impact of the ELA blend, a polyphenol-rich blend containing EP, LCK, and AM, on the cellular mechanisms involved in viral infection. To assess the effects of the ELA blend, various experiments were conducted using A549 cells and a mucociliary tissue 3D model called EpiAirway™. Inflammation and oxidative stress induced by LPS were evaluated through measurements of SOD activity, ELISA, and qPCR analysis. Additionally, antiviral assays were performed in a cell-present environment to examine the blend's effectiveness against HCoV-OC43. The results showed that the ELA blend-treated group exhibited reduced expression of IL1B, CXCL8, ICAM1, MCP1, and RELA in both A549 cells and EpiAirway™. Moreover, the blend enhanced the expression of CAT, HMOX1, SOD1, and SOD2 in A549 cells. The antiviral activity of the ELA blend was also investigated, i.e. its influence on viral replication cycle, to determine the potential as an antiviral preparation. At the highest non-cytotoxic concentration, the ELA blend demonstrated a 87.5 % reduction in viral titer when administered simultaneously with HCoV-OC43. It emphasize potential ability of the preparation to block viral entry to the host cells. At the same time, ELA blend did not express virucidal activity, i.e. inactivation of free viral particles, against HCoV-OC43. In conclusion, ELA blend displayed antiviral activity and exhibited immunomodulatory and antioxidant effects. Based on these findings, it can be concluded that ELA blend has potential for the prevention and treatment of viral infections.
Collapse
Affiliation(s)
- Katarzyna Zima
- AronPharma Ltd. R&D Department, Trzy Lipy Street 3, 80-172, Gdańsk, Poland
- Department of Physiology, Medical University of Gdańsk, Dębinki 1 Street, 80-211, Gdańsk, Poland
| | - Barbara Khaidakov
- AronPharma Ltd. R&D Department, Trzy Lipy Street 3, 80-172, Gdańsk, Poland
| | - Marta Sochocka
- Laboratory of Virology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12 Street, 53-114, Wrocław, Poland
| | - Michał Ochnik
- Laboratory of Virology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12 Street, 53-114, Wrocław, Poland
| | - Krzysztof Lemke
- AronPharma Ltd. R&D Department, Trzy Lipy Street 3, 80-172, Gdańsk, Poland
| | - Paulina Kowalczyk
- AronPharma Ltd. R&D Department, Trzy Lipy Street 3, 80-172, Gdańsk, Poland
- 3P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7 Street, 80-211, Gdańsk, Poland
| |
Collapse
|
463
|
Hashem A, Khalouf A, Mohamed MS, Nayfeh T, Elkhapery A, Zahid S, Altibi A, Thyagaturu H, Kashou A, Anavekar NS, Gulati M, Balla S. Racial, ethnic and sex disparity in acute heart failure patients with COVID-19: A nationwide analysis. Heliyon 2024; 10:e34513. [PMID: 39157311 PMCID: PMC11327804 DOI: 10.1016/j.heliyon.2024.e34513] [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: 11/16/2023] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 08/20/2024] Open
Abstract
Background Patients with acute heart failure (AHF) exacerbation are susceptible to complications in the setting of COVID-19 infection. Data regarding the racial/ethnic and sex disparities in patients with AHF and COVID-19 remains limited. Objective We aim to evaluate the impact of race, ethnicity, and sex on the in-hospital outcomes of AHF with COVID-19 infection using the data from the National Inpatient Sample (NIS). Methods We extracted data from the NIS (2020) by using ICD-10-CM to identify all hospitalizations with a diagnosis of AHF and COVID-19 in the year 2020. The associations between sex, race/ethnicity, and outcomes were examined using a multivariable logistic regression model. Results We identified a total of 158,530 weighted AHF hospitalizations with COVID-19 infection in 2020. The majority were White (63.9 %), 23.3 % were Black race, and 12.8 % were of Hispanic ethnicity, mostly males (n = 84,870 [53.5 %]). After adjustment, the odds of in-hospital mortality were lowest in White females (aOR 0.83, [0.78-0.98]) and highest in Hispanic males (aOR 1.27 [1.13-1.42]) compared with White males. Overall, the odds of cardiac arrest (aOR 1.54 [1.27-1.85]) and AKI (aOR 1.36 [1.26-1.47] were higher, while odds for procedural interventions such as PCI (aOR 0.23 [0.10-0.55]), and placement on a ventilator (aOR 0.85 [0.75-0.97]) were lower among Black males in comparison to White males. Conclusion Male sex was associated with a higher risk of in-hospital mortality in white and black racial groups, while no such association was noted in the Hispanic group. Hispanic males had the highest odds of death compared with White males.
Collapse
Affiliation(s)
- Anas Hashem
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Amani Khalouf
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Mohamed Salah Mohamed
- Department of Cardiovascular Medicine, Allegheny General Hospital, Pittsburg, PA, USA
| | - Tarek Nayfeh
- Evidence-based Medicine, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Ahmed Elkhapery
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Salman Zahid
- Department of Cardiovascular Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ahmed Altibi
- Electrophysiology & Cardiac Arrhythmia Program, Department of Cardiovascular Medicine, Yale-New Haven Hospital, New Haven, CT, USA
| | - Harshith Thyagaturu
- Department of Cardiovascular Medicine, West Virginia University, Morgantown, WV, USA
| | - Anthony Kashou
- Department of Cardiovascular Medicine, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Nandan S Anavekar
- Department of Cardiovascular Medicine, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Martha Gulati
- Department of Preventive Cardiology, Barbra Streisand Women's Heart Center, Cedars Siani, Los Angeles, CA, USA
| | - Sudarshan Balla
- Department of Cardiovascular Medicine, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
464
|
Mundhara N, Sadhukhan P. Cracking the Codes behind Cancer Cells' Immune Evasion. Int J Mol Sci 2024; 25:8899. [PMID: 39201585 PMCID: PMC11354234 DOI: 10.3390/ijms25168899] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Immune evasion is a key phenomenon in understanding tumor recurrence, metastasis, and other critical steps in tumor progression. The tumor microenvironment (TME) is in constant flux due to the tumor's ability to release signals that affect it, while immune cells within it can impact cancer cell behavior. Cancer cells undergo several changes, which can change the enrichment of different immune cells and modulate the activity of existing immune cells in the tumor microenvironment. Cancer cells can evade immune surveillance by downregulating antigen presentation or expressing immune checkpoint molecules. High levels of tumor-infiltrating lymphocytes (TILs) correlate with better outcomes, and robust immune responses can control tumor growth. On the contrary, increased enrichment of Tregs, myeloid-derived suppressor cells, and M2-like anti-inflammatory macrophages can hinder effective immune surveillance and predict poor prognosis. Overall, understanding these immune evasion mechanisms guides therapeutic strategies. Researchers aim to modulate the TME to enhance immune surveillance and improve patient outcomes. In this review article, we strive to summarize the composition of the tumor immune microenvironment, factors affecting the tumor immune microenvironment (TIME), and different therapeutic modalities targeting the immune cells. This review is a first-hand reference to understand the basics of immune surveillance and immune evasion.
Collapse
Affiliation(s)
| | - Pritam Sadhukhan
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
465
|
Lazarou E, Koutsianas C, Vlachakis PK, Theofilis P, Vassilopoulos D, Tsioufis C, Lazaros G, Tousoulis D. Novel Treatments in Refractory Recurrent Pericarditis. Pharmaceuticals (Basel) 2024; 17:1069. [PMID: 39204174 PMCID: PMC11357540 DOI: 10.3390/ph17081069] [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: 07/22/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Refractory recurrent pericarditis is a troublesome condition that severely impairs the quality of life of affected patients and significantly increases healthcare spending. Until recently, therapeutic options included only a few medications and most of the patients resorted to chronic glucocorticoid treatment with steroid dependence. In the most recent decade, the introduction of interleukin-1 blockers in clinical practice has revolutionized the treatment of glucocorticoid-dependent and colchicine-resistant recurrent pericarditis due to their excellent efficacy and good safety profile. The rationale for the introduction of this class of medications in clinical practice is the autoinflammatory nature of recurrent pericarditis in a substantial rate of cases, with interleukin-1 being the main pro-inflammatory cytokine involved in this context. This review aims to discuss the contemporary available evidence from original research and real-world data on interleukin-1 blocker use in refractory recurrent pericarditis, in terms of indications, mechanism of action, efficacy, side effects, and recommended treatment protocols. Moreover, novel treatment proposals, such as hydroxychloroquine, beta blockers, and cannabidiol, which showed encouraging preliminary results, are addressed. Finally, gaps in knowledge, unmet needs, and future perspectives related to recurrent pericarditis are thoroughly discussed.
Collapse
Affiliation(s)
- Emilia Lazarou
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Vas. Sofias 114, 11527 Athens, Greece; (E.L.); (P.K.V.); (P.T.); (C.T.); (G.L.)
| | - Christos Koutsianas
- Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 114 Vass. Sophias Ave, 11527 Athens, Greece; (C.K.); (D.V.)
| | - Panayotis K. Vlachakis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Vas. Sofias 114, 11527 Athens, Greece; (E.L.); (P.K.V.); (P.T.); (C.T.); (G.L.)
| | - Panagiotis Theofilis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Vas. Sofias 114, 11527 Athens, Greece; (E.L.); (P.K.V.); (P.T.); (C.T.); (G.L.)
| | - Dimitrios Vassilopoulos
- Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 114 Vass. Sophias Ave, 11527 Athens, Greece; (C.K.); (D.V.)
| | - Costas Tsioufis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Vas. Sofias 114, 11527 Athens, Greece; (E.L.); (P.K.V.); (P.T.); (C.T.); (G.L.)
| | - George Lazaros
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Vas. Sofias 114, 11527 Athens, Greece; (E.L.); (P.K.V.); (P.T.); (C.T.); (G.L.)
| | - Dimitris Tousoulis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Vas. Sofias 114, 11527 Athens, Greece; (E.L.); (P.K.V.); (P.T.); (C.T.); (G.L.)
| |
Collapse
|
466
|
Qiu X, Ma T, Zhao S, Zheng Z. Causality between herpes virus infections and allograft dysfunction after tissue and organ transplantation: a two-sample bidirectional Mendelian randomization study. Front Immunol 2024; 15:1411771. [PMID: 39211049 PMCID: PMC11359570 DOI: 10.3389/fimmu.2024.1411771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Background Observational studies have suggested that herpes virus infections increase the risk of allograft dysfunction after tissue and organ transplantation, but it is still unclear whether this association is causal. The aim of this study was to assess the causal relationship between four herpes virus infections and allograft dysfunction. Methods We used two-sample bidirectional Mendelian randomization (MR) to investigate the causality between four herpes virus infections - cytomegalovirus (CMV), Epstein-Barr virus (EBV), herpes simplex virus (HSV) and varicella zoster virus (VZV) - and allograft dysfunction after tissue and organ transplantation. Based on summary data extracted from genome-wide association studies (GWAS), we chose eligible single nucleotide polymorphisms (SNPs) as instrumental variables. The Inverse variance weighted (IVW) method was used as the main analysis method, supplemented by Weighted median and MR-Egger analyses. The MR-PRESSO test, MR-Egger intercept test, heterogeneity test, leave-one-out analysis and funnel plot were used to analyze the sensitivity of MR results. Results We found EBV early antigen-D (EA-D) antibody levels and shingles were the only two variables associated with an increased risk of allograft dysfunction. No evidence of allograft dysfunction increasing the risk of the four herpes virus infections was observed. Sensitivity analyses confirmed the robustness of our results. Conclusions Our results suggest that EBV and VZV are involved in graft rejection or dysfunction. However, the relationship between CMV and HSV infections and allograft dysfunction remains unclear and requires further clarification.
Collapse
Affiliation(s)
- Xiaojuan Qiu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
- College of Mathematics, Jilin University, Changchun, China
| | - Tianjiao Ma
- Department of Rheumatology and Immunology, China-Japan Union Hospital of the Jilin University, Changchun, China
| | - Shishun Zhao
- College of Mathematics, Jilin University, Changchun, China
| | - Zongyu Zheng
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
467
|
Rockey NC, Le Sage V, Shephard M, Vargas-Maldonado N, Vu MN, Brown CA, Patel K, French AJ, Merrbach GA, Walter S, Ferreri LM, Holmes KE, VanInsberghe D, Clack HL, Prussin AJ, Lowen AC, Marr LC, Lakdawala SS. Ventilation does not affect close-range transmission of influenza virus in a ferret playpen setup. Proc Natl Acad Sci U S A 2024; 121:e2322660121. [PMID: 39361828 PMCID: PMC11331089 DOI: 10.1073/pnas.2322660121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 06/10/2024] [Indexed: 10/05/2024] Open
Abstract
Sustained community spread of influenza viruses relies on efficient person-to-person transmission. Current experimental transmission systems do not mimic environmental conditions (e.g., air exchange rates, flow patterns), host behaviors, or exposure durations relevant to real-world settings. Therefore, results from these traditional systems may not be representative of influenza virus transmission in humans. To address this pitfall, we developed a close-range transmission setup that implements a play-based scenario and used it to investigate the impact of ventilation rates on transmission. In this setup, four immunologically naive recipient ferrets were exposed to a donor ferret infected with a genetically barcoded 2009 H1N1 virus (H1N1pdm09) for 4 h. The ferrets interacted in a shared space that included toys, similar to a childcare setting. Transmission efficiency was assessed under low and high ventilation, with air exchange rates of ~1.3 h-1 and 23 h-1, respectively. Transmission efficiencies observed in three independent replicate studies were similar between ventilation conditions. The presence of infectious virus or viral RNA on surfaces and in air throughout the exposure area was also not impacted by the ventilation rate. While high viral genetic diversity in donor ferret nasal washes was maintained during infection, recipient ferret nasal washes displayed low diversity, revealing a narrow transmission bottleneck regardless of ventilation rate. Examining the frequency and duration of ferret physical touches revealed no link between these interactions and a successful transmission event. Our findings indicate that exposures characterized by frequent, close-range interactions and the presence of fomites can overcome the benefits of increased ventilation.
Collapse
Affiliation(s)
- Nicole C. Rockey
- Department of Microbiology and Molecular Genetics, The University of Pittsburgh, Pittsburgh, PA15219
| | - Valerie Le Sage
- Department of Microbiology and Molecular Genetics, The University of Pittsburgh, Pittsburgh, PA15219
| | - Meredith Shephard
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | | | - Michelle N. Vu
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | - Cambria A. Brown
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | - Krishna Patel
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | - Andrea J. French
- Department of Microbiology and Molecular Genetics, The University of Pittsburgh, Pittsburgh, PA15219
| | - Grace A. Merrbach
- Department of Microbiology and Molecular Genetics, The University of Pittsburgh, Pittsburgh, PA15219
| | - Sydney Walter
- Department of Microbiology and Molecular Genetics, The University of Pittsburgh, Pittsburgh, PA15219
| | - Lucas M. Ferreri
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | - Katie E. Holmes
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | - David VanInsberghe
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | - Herek L. Clack
- Department of Civil and Environmental Engineering, University of Michigan, Ann Arbor, MI48109
| | - Aaron J. Prussin
- Department of Civil and Environmental Engineering, Virginia Tech, Blacksburg, VA24061
| | - Anice C. Lowen
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| | - Linsey C. Marr
- Department of Civil and Environmental Engineering, Virginia Tech, Blacksburg, VA24061
| | - Seema S. Lakdawala
- Department of Microbiology and Molecular Genetics, The University of Pittsburgh, Pittsburgh, PA15219
- Department of Microbiology and Immunology, Emory University, Atlanta, GA30322
| |
Collapse
|
468
|
Djuidje Chatue IA, Nyegue MA, Kamdem SD, Maloba F, Taliy Junaid I, Malhotra P, Masumbe Netongo P. Association between Epstein-Barr virus reactivation and severe malaria in pregnant women living in a malaria-endemic region of Cameroon. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003556. [PMID: 39133703 PMCID: PMC11318859 DOI: 10.1371/journal.pgph.0003556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 07/10/2024] [Indexed: 08/15/2024]
Abstract
Malaria kills nearly 619,000 people each year. Despite the natural immunity acquired to malaria, pregnant women and children under five die from severe forms of the disease in sub-Saharan Africa. Co-infection with acute Epstein-Barr Virus (EBV) infection has been shown to suppress the anti-malarial humoral responses, but little is known about the impact of EBV reactivation on malaria-associated morbidity. This study investigated the association between EBV reactivation and malaria severity in pregnant women living in a malaria-endemic region in Cameroon. A cross-sectional study was conducted on 220 pregnant women attending antenatal consultations in three health facilities in the West region of Cameroon. Malaria was diagnosed by microscopy, and Plasmodium species were identified by Nested PCR. Plasma samples were analyzed by ELISA for the presence of EBV nuclear antigen, EBV viral capsid antigen, and EBV early antigen to determine EBV reactivation. All statistics were performed using GraphPad Prism and SPSS software. The prevalence of malaria among pregnant women was 23.2%, of which 18.6% were P. falciparum mono-infections and 4.5% mixed infections (3.6% P. falciparum and P. malariae; 0.9% P. falciparum and P. ovale). 99.5% of the women were EBV seropositive, and 13.2% had EBV reactivation. Pregnant women with reactivated EBV were more likely to develop severe malaria than pregnant women with latent EBV (OR 4.33, 95% CI 1.08-17.25, p = 0.03). The median parasitemia in pregnant women with latent EBV was lower than in those with EBV reactivation (2816 vs. 19002 parasites/μL, p = 0.02). Our study revealed that lytic reactivation of EBV may be associated with the severity of malaria in pregnant women. Suggesting that, like acute infection, EBV reactivation should be considered a risk factor for severe malaria in pregnant women in malaria-endemic regions or could serve as a hallmark of malaria severity during pregnancy. Further detailed studies are needed.
Collapse
Affiliation(s)
- Ide Armelle Djuidje Chatue
- Department of Microbiology, University of Yaounde I, Yaounde, Centre, Cameroon
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (BTC-UYI), Yaounde, Centre, Cameroon
- Malaria Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, Delhi, India
| | | | - Severin Donald Kamdem
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (BTC-UYI), Yaounde, Centre, Cameroon
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Franklin Maloba
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (BTC-UYI), Yaounde, Centre, Cameroon
- Department of Microbiology and Parasitology, University of Buea, Buea, Southwest Cameroon
- Biology Program, School of Science, Navajo Technical University, Crownpoint, New Mexico, United States of America
| | - Iqbal Taliy Junaid
- Malaria Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, Delhi, India
| | - Pawan Malhotra
- Malaria Biology, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, Delhi, India
| | - Palmer Masumbe Netongo
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (BTC-UYI), Yaounde, Centre, Cameroon
- Biology Program, School of Science, Navajo Technical University, Crownpoint, New Mexico, United States of America
- Department of Biochemistry, University of Yaounde I, Yaounde, Centre, Cameroon
| |
Collapse
|
469
|
Bhowmick S, Gupta S, Mondal S, Mallick AI. Activation of Antiviral Host Responses against Avian Influenza Virus and Remodeling of Gut Microbiota by rLAB Vector Expressing rIL-17A in Chickens. ACS Infect Dis 2024; 10:3026-3041. [PMID: 38970488 DOI: 10.1021/acsinfecdis.4c00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Low-pathogenic avian influenza virus (LPAIV) remains the most common subtype of type-A influenza virus that causes moderate to severe infection in poultry with significant zoonotic and pandemic potential. Due to high mutability, increasing drug resistance, and limited vaccine availability, the conventional means to prevent intra- or interspecies transmission of AIV is highly challenging. As an alternative to control AIV infections, cytokine-based approaches to augment antiviral host defense have gained significant attention. However, the selective application of cytokines is critical since unregulated expression of cytokines, particularly proinflammatory ones, can cause substantial tissue damage during acute phases of immune responses. Moreover, depending on the type of cytokine and its impact on intestinal microbiota, outcomes of cytokine-gut microflora interaction can have a critical effect on overall host defense against AIV infections. Our recent study demonstrated some prominent roles of chicken IL-17A (ChIL-17A) in regulating antiviral host responses against AIV infection, however, in an in vitro model. For more detailed insights into ChIL-17A function, in the present study, we investigated whether ChIL-17A-meditated elevated antiviral host responses can translate into effective immune protection against AIV infection in an in vivo system. Moreover, considering the role of gut health in fostering innate or local host responses, we further studied the contributory relationships between gut microbiota and host immunity against AIV infection in chickens. For this, we employed a recombinant lactic acid-producing bacterial (LAB) vector, Lactococcus lactis, expressing ChIL-17A and analyzed the in vivo functionality in chickens against an LPAIV (A/H9N2) infection. Our study delineates that mucosal delivery of rL. lactis expressing ChIL-17A triggers proinflammatory signaling cascades and can drive a positive shift in phylum Firmicutes, along with a marked decline in phylum Actinobacteriota and Proteobacteria, favoring effective antiviral host responses against AIV infection in chickens. We propose that ChIL-17A-mediated selective expansion of beneficial gut microbiota might form a healthy microbial community that augments the effective immune protection against AIV infections in chickens.
Collapse
Affiliation(s)
- Sucharita Bhowmick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Subhadeep Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Samiran Mondal
- Department of Veterinary Pathology, West Bengal University of Animal and Fishery Sciences, Kolkata 700037, West Bengal, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| |
Collapse
|
470
|
Li S, Xie Y, Yu C, Zheng C, Xu Z. The battle between host antiviral innate immunity and immune evasion by cytomegalovirus. Cell Mol Life Sci 2024; 81:341. [PMID: 39120730 PMCID: PMC11335264 DOI: 10.1007/s00018-024-05369-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Cytomegalovirus (CMV) has successfully established a long-lasting latent infection in humans due to its ability to counteract the host antiviral innate immune response. During coevolution with the host, the virus has evolved various evasion techniques to evade the host's innate immune surveillance. At present, there is still no vaccine available for the prevention and treatment of CMV infection, and the interaction between CMV infection and host antiviral innate immunity is still not well understood. However, ongoing studies will offer new insights into how to treat and prevent CMV infection and its related diseases. Here, we update recent studies on how CMV evades antiviral innate immunity, with a focus on how CMV proteins target and disrupt critical adaptors of antiviral innate immune signaling pathways. This review also discusses some classic intrinsic cellular defences that are crucial to the fight against viral invasion. A comprehensive review of the evasion mechanisms of antiviral innate immunity by CMV will help investigators identify new therapeutic targets and develop vaccines against CMV infection.
Collapse
Affiliation(s)
- Shuang Li
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuanyang Xie
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
471
|
Cheng H, Jiao L, Liu B. The influence of COVID-19 pandemic on body mass and cardiopulmonary endurance of Chinese adolescents: a longitudinal follow-up study. Front Public Health 2024; 12:1406120. [PMID: 39171303 PMCID: PMC11335510 DOI: 10.3389/fpubh.2024.1406120] [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: 03/24/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND With the spread and spread of COVID-19 around the world, youth's learning, lifestyle and health have been greatly affected. Based on the current research, there is no adequate analysis of the development of young people's physique and heart and lung health during COVID-19, and there is a lack of relevant targeted research. The aim of this study was to investigate the changes of BMI and Maximum Oxygen Absorption (VO2max) in 12-14 year old teenagers before and after COVID-19. METHOD The BMI, 1,000/800 m running time and associated data related to 29,813 individuals between 2019 and 2022 were collected by cluster sampling, and the changes of BMI Z and VO2max before and after the outbreak were analyzed. Moreover, the relationship between BMI and cardiovascular endurance was analyzed by means of multi-linear stepwise regression. RESULTS The covariance analysis models indicated that compared with 2019, adolescent weight, BMI, and 1,000/800 m running time showed varying degrees of growth in 2020, while lung capacity decreased. All indicators achieved rapid rebound in 2021 and 2022 (p < 0.01); the one-way analysis of variance models indicated that The BMI Z score and VO2max of adolescents showed growth and decline in 2020, respectively, and achieved rapid recovery and development in 2021 and 2022 (p < 0.01). The results of the multiple linear stepwise regression analysis indicate that, after the years of BMI Z and novel coronavirus infection were included (△R2 = 0.179), adolescents' overweight and obesity were positively correlated with the maximum oxygen uptake (B = 0.643, 95%CI = 0.634 ~ 0.652); There is a negative correlation between weight loss and maximum oxygen uptake (B = -0.510, 95%CI = -0.537~-0.484); The year of novel coronavirus infection was positively correlated with the maximum oxygen uptake of adolescents (B = 0.116, 95%CI = 0.107~0.125). CONCLUSION This study shows that the impact of COVID-19 on BMI and heart and lung health in adolescents is significant. Young people of all ages and sexes showed similar developmental trends.
Collapse
Affiliation(s)
- Hao Cheng
- College of Physical Education, Shandong Sport University, Jinan, Shandong, China
| | - Long Jiao
- Basic Education Group, Shandong Normal University, Jinan, Shandong, China
| | - Bin Liu
- Physical Education Teaching and Research Group, Jinan Licheng No.2 High School, Jinan, Shandong, China
| |
Collapse
|
472
|
Prudhomme J, Depaquit J, Robert-Gangneux F. Phlebotomine sand fly distribution and abundance in France: A systematic review. Parasite 2024; 31:45. [PMID: 39109982 PMCID: PMC11305120 DOI: 10.1051/parasite/2024045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Global changes in climate are contributing to modified Phlebotomine sand fly presence and activity, and the distribution of the pathogens they transmit (e.g., Leishmania and Phlebovirus), and are leading to their possible extension toward northern France. To predict the evolution of these pathogens and control their spread, it is essential to identify and characterize the presence and abundance of potential vectors. However, there are no recent publications describing sand fly species distribution in France. Consequently, we carried out a systematic review to provide distribution and abundance maps over time, along with a simplified dichotomous key for species in France. The review adhered to PRISMA guidelines, resulting in 172 relevant capture reports from 168 studies out of the 2646 documents retrieved, of which 552 were read and 228 analyzed. Seven species were recorded and categorized into three groups based on their abundance: low abundance species, abundant but little-studied species, and abundant vector species. Sand flies are certainly present throughout France but there is a greater diversity of species in the Mediterranean region. Phlebotomus perniciosus and Ph. ariasi are the most abundant and widely distributed species, playing a role as vectors of Leishmania. Sergentomyia minuta, though very abundant, remains under-studied, highlighting the need for further research. Phlebotomus papatasi, Ph. perfiliewi, Ph. sergenti, and Ph. mascittii are present in low numbers and are less documented, limiting understanding of their potential role as vectors. This work provides the necessary basis for comparison of field data generated in the future.
Collapse
Affiliation(s)
- Jorian Prudhomme
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement Travail), UMR_S 1085 35000 Rennes France
| | - Jérôme Depaquit
- Université de Reims Champagne-Ardenne, Faculté de Pharmacie, UR EpidémioSurveillance et Circulation de Parasites dans les Environnements (ESCAPE), and ANSES, USC Pathogènes-Environnement-Toxoplasme-Arthropodes-Réservoirs-bioDiversité (PETARD) Reims France
- Centre Hospitalo-Universitaire, Laboratoire de Parasitologie-Mycologie 51092 Reims France
| | - Florence Robert-Gangneux
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement Travail), UMR_S 1085 35000 Rennes France
| |
Collapse
|
473
|
Petrea Cliveți CL, Ciortea DA, Miulescu M, Candussi IL, Chirila SI, Verga Răuță GI, Bergheș SE, Râșcu MC, Berbece SI. A New Case of Paediatric Systemic Lupus Erythematosus with Onset after SARS-CoV-2 and Epstein-Barr Infection-A Case Report and Literature Review. Curr Issues Mol Biol 2024; 46:8642-8657. [PMID: 39194726 DOI: 10.3390/cimb46080509] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Viral infections caused by exposure to viruses such as Epstein-Barr, cytomegalovirus, or Parvovirus B19 have always been considered predisposing environmental factors for the onset of autoimmune diseases. More recently, autoimmune mechanisms such as molecular mimicry, T-cell activation, transient immunosuppression and inflammation have also been observed in cases of SARS-CoV-2 infection. Several newly diagnosed autoimmune disorders have been reported post-COVID-19, such as COVID-19-associated multisystemic inflammatory syndrome in children (MIS-C), type 1 diabetes mellitus, systemic lupus erythematosus, or rheumatoid arthritis. In this article, we present a new case of paediatric systemic lupus erythematosus (SLE) with haematological (macrophage activation syndrome), renal (stage 2), cutaneous (urticarial vasculitis) and digestive involvement, onset three and a half months post-COVID-19. In the dynamics, de novo infection generated by Epstein-Barr exposure was associated. The diagnosis was confirmed based on EULAR/ACR 2019 criteria. The aim of the article is to present a possible correlation between SARS-CoV-2 and Epstein-Barr as extrinsic factors in triggering or activating paediatric systemic lupus erythematosus. Keywords: paediatric systemic lupus erythematosus; post-COVID-19; Epstein-Barr; SARS- CoV-2; case report; paediatric patient.
Collapse
Affiliation(s)
- Carmen Loredana Petrea Cliveți
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos" of Galati, 800008 Galati, Romania
- Emergency Clinical Hospital for Children "Sf Ioan", 800487 Galati, Romania
| | - Diana-Andreea Ciortea
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos" of Galati, 800008 Galati, Romania
- Emergency Clinical Hospital for Children "Maria Sklodowska Curie", 041451 Bucharest, Romania
| | - Magdalena Miulescu
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos" of Galati, 800008 Galati, Romania
- Emergency Clinical Hospital for Children "Sf Ioan", 800487 Galati, Romania
| | - Iuliana-Laura Candussi
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos" of Galati, 800008 Galati, Romania
- Emergency Clinical Hospital for Children "Sf Ioan", 800487 Galati, Romania
| | | | - Gabriela Isabela Verga Răuță
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos" of Galati, 800008 Galati, Romania
- Emergency Clinical Hospital for Children "Sf Ioan", 800487 Galati, Romania
| | | | - Mihai Ciprian Râșcu
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos" of Galati, 800008 Galati, Romania
| | - Sorin Ion Berbece
- Faculty of Medicine and Pharmacy, University "Dunarea de Jos" of Galati, 800008 Galati, Romania
| |
Collapse
|
474
|
Cui L, Li X, Chen Z, Liu Z, Zhang Y, Han Z, Liu S, Li H. Integrative RNA-seq and ChIP-seq analysis unveils metabolic regulation as a conserved antiviral mechanism of chicken p53. Microbiol Spectr 2024; 12:e0030924. [PMID: 38888361 PMCID: PMC11302347 DOI: 10.1128/spectrum.00309-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/02/2024] [Indexed: 06/20/2024] Open
Abstract
The tumor suppressor p53, primarily functioning as a transcription factor, has exhibited antiviral capabilities against various viruses in chickens, including infectious bursal disease virus (IBDV), avian leukosis virus subgroup J (ALV-J), and avian infectious laryngotracheitis virus (ILTV). Nevertheless, the existence of a universal antiviral mechanism employed by chicken p53 (chp53) against these viruses remains uncertain. This study conducted a comprehensive comparison of molecular networks involved in chp53's antiviral function against IBDV, ALV-J, and ILTV. This was achieved through an integrated analysis of ChIP-seq data, examining chp53's genome-wide chromatin occupancy, and RNA-seq data from chicken cells infected with these viruses. The consistent observation of chp53 target gene enrichment in metabolic pathways, confirmed via ChIP-qPCR, suggests a ubiquitous regulation of host cellular metabolism by chp53 across different viruses. Further genome binding motif conservation analysis and transcriptional co-factor prediction suggest conserved transcriptional regulation mechanism by which chp53 regulates host cellular metabolism during viral infection. These findings offer novel insights into the antiviral role of chp53 and propose that targeting the virus-host metabolic interaction through regulating p53 could serve as a universal strategy for antiviral therapies in chickens.IMPORTANCEThe current study conducted a comprehensive analysis, comparing molecular networks underlying chp53's antiviral role against infectious bursal disease virus (IBDV), avian leukosis virus subgroup J (ALV-J), and avian infectious laryngotracheitis virus (ILTV). This was achieved through a combined assessment of ChIP-seq and RNA-seq data obtained from infected chicken cells. Notably, enrichment of chp53 target genes in metabolic pathways was consistently observed across viral infections, indicating a universal role of chp53 in regulating cellular metabolism during diverse viral infections. These findings offer novel insights into the antiviral capabilities of chicken p53, laying a foundation for the potential development of broad-spectrum antiviral therapies in chickens.
Collapse
Affiliation(s)
- Lu Cui
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuefeng Li
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhijie Chen
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zheyi Liu
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yu Zhang
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zongxi Han
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shengwang Liu
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hai Li
- Division of Avian Infectious Diseases, State Key Laboratory for Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
475
|
Chai H, Wei Y, Chen W, Han G, Godspower BO, Liu Y, Dong C, Zhang Z, Li Y. Protection efficacy and the safety of the synergy between modified Bazhen powder and PRRSV modified-live virus vaccine against HP-PRRSV in piglets. Front Vet Sci 2024; 11:1436426. [PMID: 39161459 PMCID: PMC11331794 DOI: 10.3389/fvets.2024.1436426] [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: 05/22/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024] Open
Abstract
The highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) poses a significant threat to the global swine industry. Vaccination is a preventive measure against viral infections. However, the use of vaccines in livestock healthcare programs faces the challenge of safety and delayed immune responses. Earlier studies have shown the potential of modified Bazhen powder as an immunomodulator with significant biological properties, but its effect on vaccines against HP-PRRSV is yet to be studied. This study elucidated how modified Bazhen powder could improve the safety and efficacy of the conventional PRRSV vaccine by evaluating T-cell responses, antibody levels, clinical symptoms, levels of viremia, organ health, and cytokine production. The results revealed that the oral application of modified Bazhen powder in combination with PRRS vaccination improved both cellular and humoral immunity, accelerated viremia clearance, improved lung injury scores, and reduced viral load in the tonsils. The modified Bazhen powder also effectively reduced inflammatory responses following a PRRSV challenge. These findings further highlight the pharmacological properties of modified Bazhen powder as a potential oral immunomodulatory agent that could enhance vaccine efficacy and ensure broad-spectrum protection against HP-PRRSV in pigs.
Collapse
Affiliation(s)
- Hua Chai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanru Wei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wenguang Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Guorui Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Bello-Onaghise Godspower
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Department of Animal Science, Faculty of Agriculture, University of Benin, Benin City, Nigeria
| | - Yanyan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chunliu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhiyun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
476
|
Danila AI, Cioca F, Gadde ST, Daruvuri SP, Timar R, Hogea E. Prognostic Utility of dNLR, ALRI, APRI, and SII in COVID-19 Patients with Diabetes: A Cross-Sectional Study. Diagnostics (Basel) 2024; 14:1685. [PMID: 39125561 PMCID: PMC11311620 DOI: 10.3390/diagnostics14151685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has necessitated the identification of biomarkers that can predict disease severity, particularly in vulnerable populations such as individuals with diabetes. This study aims to evaluate the predictive value of inflammatory and liver function markers, specifically derived Neutrophil to Lymphocyte Ratio (dNLR), aspartate aminotransferase (AST)-to-lymphocyte ratio (ALRI), AST to Platelet Ratio Index (APRI), and Systemic Inflammation Index (SII), in COVID-19 patients with and without diabetes. This cross-sectional study included 336 participants, comprising 168 patients with diabetes matched with 168 without, based on gender, body mass index (BMI), and COVID-19 severity at hospitalization. The study was conducted at Victor Babes Hospital for Infectious Diseases and Pulmonology from January 2021 to December 2023. All participants had a confirmed SARS-CoV-2 infection and met the inclusion criteria of being 18 years or older with type 1 or type 2 diabetes as per American Diabetes Association guidelines. At 3 days post symptom onset, significant differences in inflammatory and liver function markers were observed between the two groups. The dNLR, ALRI, APRI, and SII were notably higher in diabetic patients. At a dNLR cutoff of 2.685, the sensitivity and specificity were 70.312% and 65.978%, respectively, with an AUC of 0.624 (p < 0.001). The ALRI showed a cutoff of 0.812, with a sensitivity of 76.429% and specificity of 69.541% (AUC 0.752, p < 0.001). These markers demonstrated statistically significant hazard ratios at both 3 and 7 days, indicating their predictive relevance for severe COVID-19 outcomes. For instance, at 7 days, SII demonstrated a hazard ratio of 2.62 (CI: 1.29-5.04, p < 0.001), highlighting its strong prognostic capability. The study successfully identified significant differences in inflammatory and liver function markers between COVID-19 patients with and without diabetes, with these markers showing good predictive value for disease severity. The results underscore the potential of these biomarkers, particularly ALRI and SII, as valuable tools in managing COVID-19, aiding in the timely identification of patients at increased risk of severe outcomes.
Collapse
Affiliation(s)
- Alexandra Ioana Danila
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
- Department of Anatomy and Embryology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Flavius Cioca
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
- Discipline of Medical Statistics and Bioinformatics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Sai Teja Gadde
- Faculty of General Medicine, All India Institute of Medical Sciences (AIIMS), Mangalagiri 522503, India;
| | - Sai Praveen Daruvuri
- Faculty of General Medicine, Bukovinian State Medical University, Teatralna Square, 2, 58002 Chernivtsi, Ukraine;
| | - Romulus Timar
- Department of Internal Medicine II, Division of Diabetes, Nutrition and Metabolic Diseases, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Elena Hogea
- Discipline of Microbiology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| |
Collapse
|
477
|
Mostafavi E, Ebrahimi B, Doosti-Irani A, Mirzazadeh A. Prevalence of hepatitis B, hepatitis C, and tuberculosis among people living with HIV in Iran: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:777. [PMID: 39097724 PMCID: PMC11297631 DOI: 10.1186/s12879-024-09676-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Up to now several studies estimate the prevalence of HBV, HCV, and TB among people living with HIV (PLWH) in Iran; however, their results are inconsistent. This study aimed to estimate the overall prevalence of HBV, HVC, and TB among Iranian PLWH. METHODS In this systematic review and meta-analysis six databases including Medline, Web of Science, Scopus, MagIran, Scientific Information Database (SID), and Barakat Knowledge network system were searched up to October 2023 with no language restriction. All studies estimated the prevalence of HBV, HCV, and TB among PLWH in Iran were included. The random-effects model was used to report the study estimates. Results were reported at a 95% confidence interval (CI). RESULTS Out of 1050 retrieved references, 58 articles met the eligibility criteria. Overall among PLWH, HBV prevalence was 13.0% (95% CI: 11.0, 15.0), HCV prevalence was 54% (95% CI: 45.0, 64.0), and TB prevalence was 19% (95% CI: 13.0, 24.0). The results from multivariate meta-regression analysis showed no statistically significant association between HBV and TB prevalence with the year of study, quality of studies, age, gender, and persons who inject drugs (PWID). HCV prevalence was significantly associated with PWID. CONCLUSION We found HBV, HCV, and TB infections are common among PLWH in Iran and required to be screened and treated with effective and timely services.
Collapse
Affiliation(s)
- Ehsan Mostafavi
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Bahram Ebrahimi
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Science, Hamadan, Iran
| | - Amin Doosti-Irani
- Department of Epidemiology, School of Public Health and Research Center for Health Sciences, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Ali Mirzazadeh
- HIV/STI Surveillance Research Center, and WHO Collaborating for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
- Department of Epidemiology and Biostatistics, Institute for Global Health Sciences, University of California, San Francisco, CA, USA
| |
Collapse
|
478
|
Ortis M, Chevalier M, Olivieri CV, Vitale S, Paul A, Tonoyan L, Doglio A, Marsault R. Herpes Simplex Virus Type 1 Infection of Human Periodontal Ligament. Int J Mol Sci 2024; 25:8466. [PMID: 39126036 PMCID: PMC11312683 DOI: 10.3390/ijms25158466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
The periodontal ligament (PDL) is a complex connective tissue that connects the tooth root to the dental alveolar bone and plays crucial mechanical roles. PDL also exhibits regenerative roles and regulatory functions to maintain periodontium integrity and homeostasis. While PDL exposure to oral microbial pathogens is common, virtually nothing is known regarding viral infections of PDL. In particular, human herpes simplex virus type 1 (HSV-1) persistently infects the oral cavity through infections of the oral epithelium, connective tissue and neurons. While the oral spread of HSV-1 is generally asymptomatic, this virus has also been implicated in various oral pathologies. In this study, using a primary cell model derived from PDL (PDL cells), and whole surgical fragments of PDL, we provide evidence supporting the efficient infection of PDL by HSV-1 and the promotion of cytopathic effects. Infection of PDL by HSV-1 was also associated with an acute innate inflammatory response, as illustrated by the production of antiviral interferons and pro-inflammatory cytokines. Furthermore, this inflammatory response to HSV-1 was exacerbated in the presence of bacterial-derived products, such as peptidoglycans. This work therefore highlights the ability of HSV-1 to infect mesenchymal cells from PDL, suggesting that PDL may serve as a viral reservoir for the periodontal spread of HSV-1. Moreover, this raises questions about HSV-1 oral pathogenesis, as HSV-1-associated cytopathic and inflammatory effects may contribute to profound alterations of PDL integrity and functioning.
Collapse
Affiliation(s)
- Morgane Ortis
- Laboratoire MICORALIS, Faculté de Chirurgie Dentaire, Université Côte d’Azur, 5, Rue du 22ème BCA, 06300 Nice, France; (M.O.); (M.C.); (C.-V.O.); (A.P.); (L.T.); (R.M.)
| | - Marlène Chevalier
- Laboratoire MICORALIS, Faculté de Chirurgie Dentaire, Université Côte d’Azur, 5, Rue du 22ème BCA, 06300 Nice, France; (M.O.); (M.C.); (C.-V.O.); (A.P.); (L.T.); (R.M.)
| | - Charles-Vivien Olivieri
- Laboratoire MICORALIS, Faculté de Chirurgie Dentaire, Université Côte d’Azur, 5, Rue du 22ème BCA, 06300 Nice, France; (M.O.); (M.C.); (C.-V.O.); (A.P.); (L.T.); (R.M.)
| | - Sébastien Vitale
- Laboratoire de Virologie, Centre Hospitalier Universitaire de Nice, 06003 Nice, France;
| | - Adrien Paul
- Laboratoire MICORALIS, Faculté de Chirurgie Dentaire, Université Côte d’Azur, 5, Rue du 22ème BCA, 06300 Nice, France; (M.O.); (M.C.); (C.-V.O.); (A.P.); (L.T.); (R.M.)
| | - Lilit Tonoyan
- Laboratoire MICORALIS, Faculté de Chirurgie Dentaire, Université Côte d’Azur, 5, Rue du 22ème BCA, 06300 Nice, France; (M.O.); (M.C.); (C.-V.O.); (A.P.); (L.T.); (R.M.)
| | - Alain Doglio
- Laboratoire MICORALIS, Faculté de Chirurgie Dentaire, Université Côte d’Azur, 5, Rue du 22ème BCA, 06300 Nice, France; (M.O.); (M.C.); (C.-V.O.); (A.P.); (L.T.); (R.M.)
- Unité de Thérapie Cellulaire et Génique (UTCG), Centre Hospitalier Universitaire de Nice, 06003 Nice, France
| | - Robert Marsault
- Laboratoire MICORALIS, Faculté de Chirurgie Dentaire, Université Côte d’Azur, 5, Rue du 22ème BCA, 06300 Nice, France; (M.O.); (M.C.); (C.-V.O.); (A.P.); (L.T.); (R.M.)
| |
Collapse
|
479
|
Michaels TM, Essop MF, Joseph DE. Potential Effects of Hyperglycemia on SARS-CoV-2 Entry Mechanisms in Pancreatic Beta Cells. Viruses 2024; 16:1243. [PMID: 39205219 PMCID: PMC11358987 DOI: 10.3390/v16081243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic β-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic β-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.
Collapse
Affiliation(s)
- Tara M. Michaels
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa;
| | - Danzil E. Joseph
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| |
Collapse
|
480
|
Alshoaibi IA, Al-Gamli A, Abdullah M, Abdo B, Alzanen KH, Alhakamy M, Al-Namer M, Ahmed F, Tamesh M, Mahdi W, Abdo Z, Mohammed M. Effectiveness of Ledipasvir-Sofosbuvir 12 Weeks After Hepatitis C Virus Genotype 1 Infection and the Factors Associated With Sustained Virologic Response: A Retrospective Study. Cureus 2024; 16:e68249. [PMID: 39350869 PMCID: PMC11439843 DOI: 10.7759/cureus.68249] [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] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The combination of ledipasvir and sofosbuvir (LDV/SOF) has been licensed to treat genotype 1 hepatitis C virus infection (HCV) with a 12-week regimen. However, there is scant data from Yemen regarding this combination regimen. Here, we investigate sustained virologic responses (SVR) 12 weeks after HCV treatment with LDV/SOF regimens and the factors that contribute to SVR failure. MATERIAL AND METHOD A retrospective cross-sectional study was conducted at Althora General Hospital in Ibb, Yemen, from June 1, 2019, to October 31, 2022, on 53 cases with HCV genotype 1 infection who received combined therapy of LDV/SOF and completed treatment for 12 weeks. The clinical characteristics and treatment follow-up were obtained from patient medical records. Factors associated with SVR failure were investigated in univariate analysis with odds ratio (OR) and 95% confidence interval (CI). RESULT The mean age was 50 ± 15.3 years, and most cases were female (n=36, 67.9%). Comorbidities were diabetes, hypertension, and fatty liver, which were represented in 12 (22.6%), nine (17.0%), and eight (15.1%) cases, respectively. A total of 13 (24.5%) patients had compensated liver cirrhosis, while the remaining 40 patients (75.5%) were non-cirrhotic healthy individuals. The baseline viral load (HCV RNA) was more than 800000 IU/mL in 21 patients (39.6%). Early virological response (ERV) was achieved in 51 patients (96.2%). After treatment, 46 of the patients (86.8%) achieved SVR at Week 12, while failure occurred in two patients (3.8%) and relapse occurred in five patients (9.4%). Blood liver enzymes, including alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase, returned to normal, with statistically significant improvements in non-cirrhotic healthy persons than compensated liver cirrhosis individuals (p= 0.006, 0.006, and 0.010; respectively). Factors associated with SVR failure were older age (OR:1.13; 95% CI: 1.03-1.30, p=0.009), presence of liver cirrhosis (OR: 5.48; 95% CI: 1.04-28.98, p=0.031), having diabetes (OR: 6.33; 95% CI: 1.19-37.93, p= 0.019), baseline higher viral load (OR: 2.27; 95% CI: 0.45-12.73, p<0.001), and not achieving EVR (OR:7.63; 95% CI: 3.77- 17.78, p= 0.009). CONCLUSION In this study, we found that LDV/SOF regimens are effective against HCV genotype one infection, allowing for the expansion of 12-week treatment for suitable patients in clinical settings. Additionally, older age, liver cirrhosis, diabetes, higher pretreatment viral load, and non-completion of EVR were associated with SVR failure. However, due to the small number of HCV genotype 1 infected individuals in this study, more corporate data is required to get a clear conclusion.
Collapse
Affiliation(s)
| | | | | | - Basheer Abdo
- Internal Medicine, School of Medicine, Ibb University, Ibb, YEM
| | | | | | - Mamoon Al-Namer
- Internal Medicine, School of Medicine, Ibb University, Ibb, YEM
| | - Faisal Ahmed
- Urology, School of Medicine, Ibb University, Ibb, YEM
| | - Munther Tamesh
- Pharmacy, University of Science and Technology, Ibb, YEM
| | - Wadhah Mahdi
- Pharmacy, University of Science and Technology, Ibb, YEM
| | - Zeyad Abdo
- Pharmacy, University of Science and Technology, Ibb, YEM
| | - Marwa Mohammed
- General Practice, School of Medicine, Ibb University, Ibb, YEM
| |
Collapse
|
481
|
Avanthay R, Garcia-Nicolas O, Ruggli N, Grau-Roma L, Párraga-Ros E, Summerfield A, Zimmer G. Evaluation of a novel intramuscular prime/intranasal boost vaccination strategy against influenza in the pig model. PLoS Pathog 2024; 20:e1012393. [PMID: 39116029 PMCID: PMC11309389 DOI: 10.1371/journal.ppat.1012393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Live-attenuated influenza vaccines (LAIV) offer advantages over the commonly used inactivated split influenza vaccines. However, finding the optimal balance between sufficient attenuation and immunogenicity has remained a challenge. We recently developed an alternative LAIV based on the 2009 pandemic H1N1 virus with a truncated NS1 protein and lacking PA-X protein expression (NS1(1-126)-ΔPAX). This virus showed a blunted replication and elicited a strong innate immune response. In the present study, we evaluated the efficacy of this vaccine candidate in the porcine animal model as a pertinent in vivo system. Immunization of pigs via the nasal route with the novel NS1(1-126)-ΔPAX LAIV did not cause disease and elicited a strong mucosal immune response that completely blocked replication of the homologous challenge virus in the respiratory tract. However, we observed prolonged shedding of our vaccine candidate from the upper respiratory tract. To improve LAIV safety, we developed a novel prime/boost vaccination strategy combining primary intramuscular immunization with a haemagglutinin-encoding propagation-defective vesicular stomatitis virus (VSV) replicon, followed by a secondary immunization with the NS1(1-126)-ΔPAX LAIV via the nasal route. This two-step immunization procedure significantly reduced LAIV shedding, increased the production of specific serum IgG, neutralizing antibodies, and Th1 memory cells, and resulted in sterilizing immunity against homologous virus challenge. In conclusion, our novel intramuscular prime/intranasal boost regimen interferes with virus shedding and transmission, a feature that will help combat influenza epidemics and pandemics.
Collapse
MESH Headings
- Animals
- Swine
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Injections, Intramuscular
- Administration, Intranasal
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Influenza A Virus, H1N1 Subtype/immunology
- Disease Models, Animal
- Antibodies, Viral/immunology
- Immunization, Secondary/methods
- Vaccination/methods
- Influenza, Human/prevention & control
- Influenza, Human/immunology
Collapse
Affiliation(s)
- Robin Avanthay
- Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Obdulio Garcia-Nicolas
- Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Llorenç Grau-Roma
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, COMPATH, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Ester Párraga-Ros
- Department of Anatomy and Comparative Pathology, Veterinary Faculty, University of Murcia, Murcia, Spain
| | - Artur Summerfield
- Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Gert Zimmer
- Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
482
|
Hanada K. Metabolic channeling of lipids via the contact zones between different organelles. Bioessays 2024; 46:e2400045. [PMID: 38932642 DOI: 10.1002/bies.202400045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
Various lipid transfer proteins (LTPs) mediate the inter-organelle transport of lipids. By working at membrane contact zones between donor and acceptor organelles, LTPs achieve rapid and accurate inter-organelle transfer of lipids. This article will describe the emerging paradigm that the action of LTPs at organelle contact zones generates metabolic channeling events in lipid metabolism, mainly referring to how ceramide synthesized in the endoplasmic reticulum is preferentially metabolized to sphingomyelin in the distal Golgi region, how cholesterol and phospholipids receive specific metabolic reactions in mitochondria, and how the hijacking of host LTPs by intracellular pathogens may generate new channeling-like events. In addition, the article will discuss how the function of LTPs is regulated, exemplified by a few representative LTP systems, and will briefly touch on experiments that will be necessary to establish the paradigm that LTP-mediated inter-organelle transport of lipids is one of the mechanisms of compartmentalization-based metabolic channeling events.
Collapse
Affiliation(s)
- Kentaro Hanada
- Center for Quality Management Systems, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
483
|
Chou SH, Chuang C, Juan CH, Ho YC, Liu SY, Chen L, Lin YT. Mechanisms and fitness of ceftazidime/avibactam-resistant Klebsiella pneumoniae clinical strains in Taiwan. Int J Antimicrob Agents 2024; 64:107244. [PMID: 38925227 DOI: 10.1016/j.ijantimicag.2024.107244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Carbapenem-resistant Klebsiella pneumoniae (CRKP) infection is a global public health issue, and ceftazidime/avibactam is recommended by international guidelines as the preferred treatment for KPC- and OXA-48-producing CRKP. Since its introduction in Taiwan in 2019, ceftazidime/avibactam-resistant strains have emerged. Our aim is to investigate the mechanisms of ceftazidime/avibactam resistance in CRKP in Taiwan and study their associated fitness costs. METHODS Ceftazidime/avibactam-resistant CRKP strains with exposure to ceftazidime/avibactam isolated from clinical specimens were consecutively collected at Taipei Veterans General Hospital in 2020. The serial strains exhibiting ceftazidime/avibactam-susceptible and ceftazidime/avibactam-resistant phenotypes isolated from the same patient were characterized using whole-genome sequencing and tested for their growth rates and competitive abilities. RESULTS A total of 35 ceftazidime/avibactam-resistant CRKP strains were identified, with 20 being metallo-β-lactamase producers. Ten strains harboured KPC variants, exhibiting MIC for ceftazidime/avibactam ranging from 64 to ≥256 mg/L. The 10 strains demonstrating high-level ceftazidime/avibactam resistance possessed mutated KPC variants: KPC-33 (n = 3), KPC-31 (n = 1), KPC-39 (n = 1), KPC-44 (n = 1), KPC-58 (n = 1), KPC-90 (n = 1), and two novel KPC variants. Ceftazidime/avibactam-resistant strains with KPC-33 and KPC-39 showed a significant fitness cost and lower growth rate compared to their parental strains. In contrast, ceftazidime/avibactam-resistant strains with KPC-58 and KPC-58 plus D179Y showed similar growth rates and competitive abilities compared to their parental strains. CONCLUSIONS Mutated KPC variants conferred high-level ceftazidime/avibactam resistance in Taiwan. Significant fitness costs were observed in both the ceftazidime/avibactam-resistant KPC-33 and KPC-39 strains. Despite conferring a similar level of ceftazidime/avibactam resistance, different KPC variants could entail varying degrees of fitness costs.
Collapse
Affiliation(s)
- Sheng-Hua Chou
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien Chuang
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Han Juan
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chien Ho
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Szu-Yu Liu
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Liang Chen
- Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY
| | - Yi-Tsung Lin
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
484
|
Chuaychu SB, Sirisereewan C, Techakriengkrai N, Tummaruk P, Thanawongnuwech R, Nedumpun T. Enhancement of systemic virus-specific T lymphocyte responses in pigs supplemented with algae-derived β-glucan. Vet J 2024; 306:106182. [PMID: 38897378 DOI: 10.1016/j.tvjl.2024.106182] [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: 05/03/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
Algae-derived β-glucan has been widely used as a feed additive in the swine industry. The supplementation of β-glucan aims to improve growth performance and modulate the immunity of pigs. However, the potential effects of supplementing β-glucan from algae on immune responses in pigs-specifically antigen-specific immunity-must be determined. In this study, the effects of algae-derived β-glucan supplementation on growth performance, virus neutralising antibody and virus-specific T lymphocytes responses were investigated in pigs. Piglets (n=112 per treatment) were assigned to three treatments including non-supplemented group (control), β-glucan 100 g/ton supplemented group (BG100), and β-glucan 200 g/ton supplemented group (BG200). In this study, production performance of pigs was not found to be different between the experimental groups. Pigs supplemented with β-glucan exhibited high levels of classical swine fever virus (CSFV)-specific producing T lymphocytes and neutralising antibody titer, compared to the control group. Interestingly, supplementation of β-glucan significantly enhanced porcine reproductive and respiratory syndrome virus (PRRSV)-specific interferon-gamma (IFN-γ) producing T lymphocytes, including CD4+, CD8+, and CD4+CD8+ T lymphocyte subpopulations. Moreover, PRRS modified live vaccine (MLV) viremia was reduced in earlier for β-glucan-supplemented pigs compared to the control group. The findings indicate that the algae-derived β-glucan possesses biological potential as an immunomodulatory substance to enhance antiviral immunity, which may contribute to disease resistance in pigs.
Collapse
Affiliation(s)
- Sh B Chuaychu
- International Graduate Program of Veterinary Science and Technology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - C Sirisereewan
- Graduate Program in Veterinary Pathobiology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand; Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - N Techakriengkrai
- Department of Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - P Tummaruk
- Department of Obstetrics Gynaecology and Reproduction, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - R Thanawongnuwech
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - T Nedumpun
- Department of Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand.
| |
Collapse
|
485
|
Schwickert KK, Glitscher M, Bender D, Benz NI, Murra R, Schwickert K, Pfalzgraf S, Schirmeister T, Hellmich UA, Hildt E. Zika virus replication is impaired by a selective agonist of the TRPML2 ion channel. Antiviral Res 2024; 228:105940. [PMID: 38901736 DOI: 10.1016/j.antiviral.2024.105940] [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: 01/31/2024] [Revised: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
The flavivirus genus includes human pathogenic viruses such as Dengue (DENV), West Nile (WNV) and Zika virus (ZIKV) posing a global health threat due to limited treatment options. Host ion channels are crucial for various viral life cycle stages, but their potential as targets for antivirals is often not fully realized due to the lack of selective modulators. Here, we observe that treatment with ML2-SA1, an agonist for the human endolysosomal cation channel TRPML2, impairs ZIKV replication. Upon ML2-SA1 treatment, levels of intracellular genomes and number of released virus particles of two different ZIKV isolates were significantly reduced and cells displayed enlarged vesicular structures and multivesicular bodies with ZIKV envelope protein accumulation. However, no increased ZIKV degradation in lysosomal compartments was observed. Rather, the antiviral effect of ML2-SA1 seemed to manifest by the compound's negative impact on genome replication. Moreover, ML2-SA1 treatment also led to intracellular cholesterol accumulation. ZIKV and many other viruses including the Orthohepevirus Hepatitis E virus (HEV) rely on the endolysosomal system and are affected by intracellular cholesterol levels to complete their life cycle. Since we observed that ML2-SA1 also negatively impacted HEV infections in vitro, this compound may harbor a broader antiviral potential through perturbing the intracellular cholesterol distribution. Besides indicating that TRPML2 may be a promising target for combatting viral infections, we uncover a tentative connection between this protein and cholesterol distribution within the context of infectious diseases.
Collapse
Affiliation(s)
- Kerstin K Schwickert
- Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University, Jena, Germany; Department of Virology, Paul-Ehrlich-Institut, 63225, Langen, Germany; Department of Chemistry, Johannes Gutenberg-University, 55122, Mainz, Germany
| | - Mirco Glitscher
- Department of Virology, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Daniela Bender
- Department of Virology, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Nuka Ivalu Benz
- Department of Virology, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Robin Murra
- Department of Virology, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Kevin Schwickert
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, 55122, Mainz, Germany
| | - Steffen Pfalzgraf
- Department of Virology, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, 55122, Mainz, Germany
| | - Ute A Hellmich
- Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University, Jena, Germany; Centre for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany; Cluster of Excellence "Balance of the Microverse", Friedrich Schiller University, Jena, Germany.
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institut, 63225, Langen, Germany.
| |
Collapse
|
486
|
Li J, Lv J, Yu F, Zhang Y, Wang Y, Yan L, Xiao Q, Li Q, Wang C, Wang X, Hou Y, Zhang F, Zhang T. Temporal changes in plasma metabolic signatures to predict immune response of antiretroviral therapy among people living with HIV. J Med Virol 2024; 96:e29798. [PMID: 39056244 DOI: 10.1002/jmv.29798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/02/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024]
Abstract
Antiretroviral therapy (ART) is an effective treatment for people living with HIV (PLHIVs), requiring an extended period to achieve immune reconstitution. Metabolic alterations induced by ART are crucial for predicting long-term therapeutic responses, yet comprehensive investigation through large-scale clinical studies is still lacking. Here, we collected plasma samples from 108 PLHIVs to the untargeted plasma metabolomics study, based on the longitudinal metabolomics design. Cross-sectional analyzes were performed at pre- and post-ART to explore the metabolic transformation induced by the therapy. Subsequently, delta values between pre- and post-ART measurements were calculated to quantify metabolic alterations. Then, the optimal set of metabolic traits and clinical signatures were further identified and applied to construct random forest model for predicting the future therapeutic responses to ART. We found distinct ART-induced metabolic transformation among PLHIVs. After confounder-adjustments, five metabolites exhibited significant associations with future immune response: tetracosatetraenoic acid (24:4n-6) (pre-ART) (odds ratio [OR]: 0.978, 95% confidence interval [CI]: 0.955~0.997), 1-(3,4-dihydroxyphenyl)-5-hydroxy-3-decanone (pre-ART) (OR: 1.298, 95% CI: 1.061~1.727), beta-PC-M6 (change) (OR: 0.967, 95% CI: 0.938~0.993), d-Galactaro-1,4-lactone (change) (OR: 1.032, 95% CI: 1.007~1.063), Annuionone C (change) (OR: 1.100, 95% CI: 1.030~1.190). The addition of plasma metabolites to clinical markers accurately predicted immune response to ART with an area under curve of 0.91. Notably, most disrupted metabolites were significantly correlated with blood lipids, suggesting that metabolic transformation might contribute to dyslipidemia among PLHIVs. This study highlights the distinct metabolic transformation post-ART among PLHIVs and reveals the potential role of metabolic transformation as key determinants of ART efficacy.
Collapse
Affiliation(s)
- Junnan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Disease, Beijing, China
| | - Jiali Lv
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fengting Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Clinical Center for HIV/AIDS, Capital Medical University, Beijing, China
| | - Yu Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuqi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liting Yan
- Clinical Center for HIV/AIDS, Capital Medical University, Beijing, China
| | - Qing Xiao
- Clinical Center for HIV/AIDS, Capital Medical University, Beijing, China
| | - Qun Li
- Clinical Center for HIV/AIDS, Capital Medical University, Beijing, China
| | - Cheng Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Disease, Beijing, China
| | - Yan Hou
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
- Clinical Research Center, Peking University, Beijing, China
| | - Fujie Zhang
- Clinical Center for HIV/AIDS, Capital Medical University, Beijing, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
487
|
Margaret MS, Melrose J. Impaired instructive and protective barrier functions of the endothelial cell glycocalyx pericellular matrix is impacted in COVID-19 disease. J Cell Mol Med 2024; 28:e70033. [PMID: 39180511 PMCID: PMC11344469 DOI: 10.1111/jcmm.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/29/2024] [Accepted: 06/18/2024] [Indexed: 08/26/2024] Open
Abstract
The aim of this study was to review the roles of endothelial cells in normal tissue function and to show how COVID-19 disease impacts on endothelial cell properties that lead to much of its associated symptomatology. This places the endothelial cell as a prominent cell type to target therapeutically in the treatment of this disorder. Advances in glycosaminoglycan analytical techniques and functional glycomics have improved glycosaminoglycan mimetics development, providing agents that can more appropriately target various aspects of the behaviour of the endothelial cell in-situ and have also provided polymers with potential to prevent viral infection. Thus, promising approaches are being developed to combat COVID-19 disease and the plethora of symptoms this disease produces. Glycosaminoglycan mimetics that improve endothelial glycocalyx boundary functions have promising properties in the prevention of viral infection, improve endothelial cell function and have disease-modifying potential. Endothelial cell integrity, forming tight junctions in cerebral cell populations in the blood-brain barrier, prevents the exposure of the central nervous system to circulating toxins and harmful chemicals, which may contribute to the troublesome brain fogging phenomena reported in cognitive processing in long COVID disease.
Collapse
Affiliation(s)
- M. Smith Margaret
- Raymond Purves Bone and Joint Research LaboratoryKolling Institute, Northern Sydney Local Health DistrictSt. LeonardsNew South WalesAustralia
- Arthropharm Australia Pharmaceuticals Pty LtdBondi JunctionSydneyNew South WalesAustralia
| | - James Melrose
- Raymond Purves Bone and Joint Research LaboratoryKolling Institute, Northern Sydney Local Health DistrictSt. LeonardsNew South WalesAustralia
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNew South WalesAustralia
- Sydney Medical SchoolNorthern, The University of SydneySydneyNew South WalesAustralia
- Faculty of Medicine and HealthThe University of Sydney, Royal North Shore HospitalSt. LeonardsNew South WalesAustralia
| |
Collapse
|
488
|
Appak O, Sayiner AA. Comparison of the NeuMoDx™ HBV quant assay and artus QS-RGQ® kit for HBV DNA quantitation in plasma samples. Diagn Microbiol Infect Dis 2024; 109:116357. [PMID: 38776663 DOI: 10.1016/j.diagmicrobio.2024.116357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
We aimed to compare the NeuMoDx HBV Assay with the artus HBV Assay using residual plasma samples and to evaluate the discordant results. The study included 200 patient samples analyzed with the NMD assay and stored at -80 °C. Samples were analyzed by artus in 2023. Discordant results were evaluated by cobas 6800 HBV DNA Test. Excellent agreement was found between both tests. Of the 100 samples that were HBV DNA negative by NMD, 93 were negative and 7 were positive by artus. With the Cobas test, 5 samples were positive. Of the100 HBV DNA positive samples detected by NMD, 99 were positive with the artus assay. This sample was also HBV DNA negative by the Cobas test. The sensitivity and specificity of NeuMoDx were found 93 % and 99 %, respectively. There was excellent qualitative agreement and strong quantitative correlation between the NeuMoDx and artus assays for HBV DNA detection and quantitation.
Collapse
Affiliation(s)
- Ozgur Appak
- Dokuz Eylul University Faculty of Medicine, Department of Medical Microbiology, Division of Medical Virology, Izmir, Turkey.
| | - Ayca Arzu Sayiner
- Dokuz Eylul University Faculty of Medicine, Department of Medical Microbiology, Division of Medical Virology, Izmir, Turkey
| |
Collapse
|
489
|
Zhi Y, Wei J, Liu Z, Zhang Q, Zhang T, Hu G. Inhibitory effects of Belamcanda extract on inflammatory response and antiviral mechanism in H9N2 Avian influenza virus: insights from in vitro and in vivo studies. Poult Sci 2024; 103:103885. [PMID: 38851182 PMCID: PMC11208944 DOI: 10.1016/j.psj.2024.103885] [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: 01/25/2024] [Revised: 05/02/2024] [Accepted: 05/18/2024] [Indexed: 06/10/2024] Open
Abstract
Avian influenza, particularly the H9N2 subtype, presents significant challenges to poultry health, underscoring the need for effective antiviral interventions. This study explores the antiviral capabilities of Belamcanda extract, a traditional Chinese medicinal herb, against H9N2 Avian influenza virus (AIV) in specific pathogen-free (SPF) chicks. Through a comprehensive approach, we evaluated the impact of the extract on cytokine modulation and crucial immunological signaling pathways, essential for understanding the host-virus interaction. Our findings demonstrate that Belamcanda extract significantly modulates the expression of key inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interleukin-1 (IL-1), interleukin-2 (IL-2), and interleukin-6 (IL-6), which are pivotal to the host's response to H9N2 AIV infection. Western blot analysis further revealed that the extract markedly reduces the expression of critical immune signaling molecules such as toll-like receptor 3 (TLR3), TIR-domain-containing adapter-inducing interferon-β (TRIF), and nuclear factor kappa B (NF-κB). These insights into the mechanisms by which Belamcanda extract influences host immune responses and hinders viral replication highlight its potential as an innovative antiviral agent for poultry health management. The study advances our comprehension of natural compounds' antiviral mechanisms and lays the groundwork for developing strategies to manage viral infections in poultry. The demonstrated ability of Belamcanda extract to modulate immune responses and inhibit viral replication establishes it as a promising candidate for future antiviral therapy development, especially in light of the need for effective treatments against evolving influenza virus strains and the critical demand for enhanced poultry health management strategies.
Collapse
Affiliation(s)
- Yan Zhi
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Jingjie Wei
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Zhenyi Liu
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Qian Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Tao Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Ge Hu
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China.
| |
Collapse
|
490
|
Rai A, Deshpande SG, Vaidya A, Shinde RK. Advancements in Immunotherapy for Breast Cancer: Mechanisms, Efficacy, and Future Directions. Cureus 2024; 16:e68351. [PMID: 39355073 PMCID: PMC11443072 DOI: 10.7759/cureus.68351] [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: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 10/03/2024] Open
Abstract
Breast cancer is a major global health challenge characterized by its diverse biological behavior and varying treatment responses. Traditional therapies, including surgery, radiation, chemotherapy, hormonal therapy, and targeted therapy, have significantly advanced breast cancer treatment but are often limited by issues such as resistance, side effects, and variable efficacy. Immunotherapy has emerged as a transformative approach, leveraging the body's immune system to target and eliminate cancer cells. This review provides a comprehensive overview of recent advancements in immunotherapy for breast cancer, detailing the mechanisms of various therapeutic strategies, including checkpoint inhibitors, monoclonal antibodies, cancer vaccines, adoptive cell therapy, and oncolytic virus therapy. We evaluate the efficacy of these approaches in different stages of breast cancer, highlighting successes and challenges encountered in clinical settings. The review also addresses the current limitations of immunotherapy, such as treatment-related adverse effects, resistance mechanisms, and issues of cost and accessibility. We discuss promising future directions, including emerging targets, combination therapies, and personalized medicine approaches. By integrating recent research and clinical trial data, this review aims to elucidate the potential of immunotherapy to revolutionize breast cancer treatment, offering insights into its future role in improving patient outcomes and shaping the landscape of oncological care.
Collapse
Affiliation(s)
- Archita Rai
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Swati G Deshpande
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashish Vaidya
- Oncology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Raju K Shinde
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
491
|
Li H, Wan L, Liu M, Ma E, Huang L, Yang Y, Li Q, Fang Y, Li J, Han B, Zhang C, Sun L, Hou X, Li H, Sun M, Qian S, Duan X, Zhao R, Yang X, Chen Y, Wu S, Zhang X, Zhang Y, Cheng G, Chen G, Gao Q, Xu J, Hou L, Wei C, Zhong H. SARS-CoV-2 spike-induced syncytia are senescent and contribute to exacerbated heart failure. PLoS Pathog 2024; 20:e1012291. [PMID: 39102426 PMCID: PMC11326701 DOI: 10.1371/journal.ppat.1012291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 08/15/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
SARS-CoV-2 spike protein (SARS-2-S) induced cell-cell fusion in uninfected cells may occur in long COVID-19 syndrome, as circulating SARS-2-S or extracellular vesicles containing SARS-2-S (S-EVs) were found to be prevalent in post-acute sequelae of COVID-19 (PASC) for up to 12 months after diagnosis. Although isolated recombinant SARS-2-S protein has been shown to increase the SASP in senescent ACE2-expressing cells, the direct linkage of SARS-2-S syncytia with senescence in the absence of virus infection and the degree to which SARS-2-S syncytia affect pathology in the setting of cardiac dysfunction are unknown. Here, we found that the senescent outcome of SARS-2-S induced syncytia exacerbated heart failure progression. We first demonstrated that syncytium formation in cells expressing SARS-2-S delivered by DNA plasmid or LNP-mRNA exhibits a senescence-like phenotype. Extracellular vesicles containing SARS-2-S (S-EVs) also confer a potent ability to form senescent syncytia without de novo synthesis of SARS-2-S. However, it is important to note that currently approved COVID-19 mRNA vaccines do not induce syncytium formation or cellular senescence. Mechanistically, SARS-2-S syncytia provoke the formation of functional MAVS aggregates, which regulate the senescence fate of SARS-2-S syncytia by TNFα. We further demonstrate that senescent SARS-2-S syncytia exhibit shrinked morphology, leading to the activation of WNK1 and impaired cardiac metabolism. In pre-existing heart failure mice, the WNK1 inhibitor WNK463, anti-syncytial drug niclosamide, and senolytic dasatinib protect the heart from exacerbated heart failure triggered by SARS-2-S. Our findings thus suggest a potential mechanism for COVID-19-mediated cardiac pathology and recommend the application of WNK1 inhibitor for therapy especially in individuals with post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Huilong Li
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luming Wan
- Beijing Institute of Biotechnology, Beijing, China
| | - Muyi Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Enhao Ma
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Linfei Huang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yilong Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Qihong Li
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yi Fang
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jingfei Li
- Beijing Institute of Biotechnology, Beijing, China
| | - Bingqing Han
- Beijing Institute of Biotechnology, Beijing, China
| | - Chang Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Lijuan Sun
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Xufeng Hou
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Haiyang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingyu Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Sichong Qian
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xuejing Duan
- Department of Pathology, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China
| | - Ruzhou Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Xuhui Zhang
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | | | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Gengye Chen
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qi Gao
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Junjie Xu
- Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
492
|
Raja K. Comments on "Oncolytic virotherapy for oral squamous cell carcinoma using replication-competent viruses". Oral Oncol 2024; 155:106897. [PMID: 38901367 DOI: 10.1016/j.oraloncology.2024.106897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Affiliation(s)
- Kannan Raja
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha Nagar, Thandalam, Chennai 602105, India.
| |
Collapse
|
493
|
DeStefano S, Fertil D, Faust M, Sadtler K. Basic immunologic study as a foundation for engineered therapeutic development. Pharmacol Res Perspect 2024; 12:e1168. [PMID: 38894611 PMCID: PMC11187943 DOI: 10.1002/prp2.1168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 06/21/2024] Open
Abstract
Bioengineering and drug delivery technologies play an important role in bridging the gap between basic scientific discovery and clinical application of therapeutics. To identify the optimal treatment, the most critical stage is to diagnose the problem. Often these two may occur simultaneously or in parallel, but in this review, we focus on bottom-up approaches in understanding basic immunologic phenomena to develop targeted therapeutics. This can be observed in several fields; here, we will focus on one of the original immunotherapy targets-cancer-and one of the more recent targets-regenerative medicine. By understanding how our immune system responds in processes such as malignancies, wound healing, and medical device implantation, we can isolate therapeutic targets for pharmacologic and bioengineered interventions.
Collapse
Affiliation(s)
- Sabrina DeStefano
- Section on Immunoengineering, National Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMarylandUSA
| | - Daphna Fertil
- Section on Immunoengineering, National Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMarylandUSA
| | - Mondreakest Faust
- Section on Immunoengineering, National Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMarylandUSA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
494
|
Fraser ME, Kucharski C, Loh Z, Hanahoe E, Fraser MJ. Design and testing of Hepatitis Delta Ribozymes for suppression of Chikungunya virus infection in cell cultures. MEDICAL RESEARCH ARCHIVES 2024; 12. [PMID: 39324067 PMCID: PMC11423935 DOI: 10.18103/mra.v12i8.5762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Chikungunya virus is an emerging pathogen with widespread distribution in regions of Africa, India, and Asia that threatens to spread into temperate climates following the introduction of its major vector, Aedes albopictus. Recent cases have been documented in Europe, the Caribbean, and the Americas. Chikungunya virus causes a disease frequently misdiagnosed as Dengue fever, with potentially life-threatening symptoms that can result in long term debilitating arthritis. There have been ongoing investigations of possible therapeutic interventions for both acute and chronic symptoms, but to date none have proven effective in reducing the severity or lasting effects of this disease. Recently, a promising vaccine candidate has received accelerated approval, indicating the importance of remedies to this emerging worldwide health threat. Nonetheless, therapeutic interventions for Chikungunya and other mosquito borne virus diseases are urgently needed yet remain elusive. The increasing risk of spread from endemic regions via human travel and commerce, coupled with the absence of a vaccine or approved therapeutic, puts a significant proportion of the world population at risk for this disease. In this report we explore the possibility of using Specific On/oFf Adapter Hepatitis Delta Virus Ribozymes as antivirals in cells infected with Chikungunya virus. The results we obtained suggest there could be some role in using these ribozyme molecules as antiviral therapies for not only Chikungunya virus, but potentially other viruses as well.
Collapse
Affiliation(s)
- Mark E Fraser
- Department of Pulmonology, Critical Care, Sleep, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Cheryl Kucharski
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| | - Zoe Loh
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710
| | - Erin Hanahoe
- Moderna, 200 Technology Square, Cambridge MA 02139
| | - Malcolm J Fraser
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| |
Collapse
|
495
|
Li P, Liu M, He WM. Integrated Transcriptomic Analysis Reveals Reciprocal Interactions between SARS-CoV-2 Infection and Multi-Organ Dysfunction, Especially the Correlation of Renal Failure and COVID-19. Life (Basel) 2024; 14:960. [PMID: 39202702 PMCID: PMC11355357 DOI: 10.3390/life14080960] [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: 06/11/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
The COVID-19 pandemic, which is caused by the SARS-CoV-2 virus, has resulted in extensive health challenges globally. While SARS-CoV-2 primarily targets the respiratory system, clinical studies have revealed that it could also affect multiple organs, including the heart, kidneys, liver, and brain, leading to severe complications. To unravel the intricate molecular interactions between the virus and host tissues, we performed an integrated transcriptomic analysis to investigate the effects of SARS-CoV-2 on various organs, with a particular focus on the relationship between renal failure and COVID-19. A comparative analysis showed that SARS-CoV-2 triggers a systemic immune response in the brain, heart, and kidney tissues, characterized by significant upregulation of cytokine and chemokine secretion, along with enhanced migration of lymphocytes and leukocytes. A weighted gene co-expression network analysis demonstrated that SARS-CoV-2 could also induce tissue-specific transcriptional profiling. More importantly, single-cell sequencing revealed that COVID-19 patients with renal failure exhibited lower metabolic activity in lung epithelial and B cells, with reduced ligand-receptor interactions, especially CD226 and ICAM, suggesting a compromised immune response. A trajectory analysis revealed that COVID-19 patients with renal failure exhibited less mature alveolar type 1 cells. Furthermore, these patients showed potential fibrosis in the hearts, liver, and lung increased extracellular matrix remodeling activities. However, there was no significant metabolic dysregulation in the liver of COVID-19 patients with renal failure. Candidate drugs prediction by Drug Signatures database and LINCS L1000 Antibody Perturbations Database underscored the importance of considering multi-organ effects in COVID-19 management and highlight potential therapeutic strategies, including targeting viral entry and replication, controlling tissue fibrosis, and alleviating inflammation.
Collapse
Affiliation(s)
- Pai Li
- Capricorn Partner, 3000 Leuven, Belgium
| | - Meng Liu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Wei-Ming He
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| |
Collapse
|
496
|
Angelova A, Barf M, Just A, Leuchs B, Rommelaere J, Ungerechts G. H-1 Parvovirus-Induced Oncolysis and Tumor Microenvironment Immune Modulation in a Novel Heterotypic Spheroid Model of Cutaneous T-Cell Lymphoma. Cancers (Basel) 2024; 16:2711. [PMID: 39123440 PMCID: PMC11311363 DOI: 10.3390/cancers16152711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The rat protoparvovirus H-1 (H-1PV) is an oncolytic virus known for its anticancer properties in laboratory models of various human tumors, including non-Hodgkin lymphomas (NHL) of B-cell origin. However, H-1PV therapeutic potential against hematological malignancies of T-cell origin remains underexplored. The aim of the present study was to conduct a pilot preclinical investigation of H-1PV-mediated oncolytic effects in cutaneous T-cell lymphoma (CTCL), a type of NHL that is urgently calling for innovative therapies. We demonstrated H-1PV productive infection and induction of oncolysis in both classically grown CTCL suspension cultures and in a novel, in vivo-relevant, heterotypic spheroid model, but not in healthy donor controls, including peripheral blood mononuclear cells (PBMCs). H-1PV-mediated oncolysis of CTCL cells was not prevented by Bcl-2 overexpression and was accompanied by increased extracellular ATP release. In CTCL spheroid co-cultures with PBMCs, increased spheroid infiltration with immune cells was detected upon co-culture treatment with the virus. In conclusion, our preclinical data show that H-1PV may hold significant potential as an ingenious viroimmunotherapeutic drug candidate against CTCL.
Collapse
Affiliation(s)
- Assia Angelova
- Clinical Cooperation Unit Virotherapy, Infection, Inflammation and Cancer Program, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.); (A.J.); (J.R.); (G.U.)
| | - Milena Barf
- Clinical Cooperation Unit Virotherapy, Infection, Inflammation and Cancer Program, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.); (A.J.); (J.R.); (G.U.)
| | - Alexandra Just
- Clinical Cooperation Unit Virotherapy, Infection, Inflammation and Cancer Program, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.); (A.J.); (J.R.); (G.U.)
| | - Barbara Leuchs
- Biopharmaceutical Production and Development Unit, German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Jean Rommelaere
- Clinical Cooperation Unit Virotherapy, Infection, Inflammation and Cancer Program, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.); (A.J.); (J.R.); (G.U.)
| | - Guy Ungerechts
- Clinical Cooperation Unit Virotherapy, Infection, Inflammation and Cancer Program, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.); (A.J.); (J.R.); (G.U.)
- Department of Medical Oncology, National Center for Tumor Diseases Heidelberg and Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|
497
|
Pandarangga P, Doan PTK, Tearle R, Low WY, Ren Y, Nguyen HTH, Dharmayanti NI, Hemmatzadeh F. mRNA Profiling and Transcriptomics Analysis of Chickens Received Newcastle Disease Virus Genotype II and Genotype VII Vaccines. Pathogens 2024; 13:638. [PMID: 39204239 PMCID: PMC11357267 DOI: 10.3390/pathogens13080638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Newcastle Disease Virus (NDV) genotype VII (GVII) is becoming the predominant strain of NDV in the poultry industry. It causes high mortality even in vaccinated chickens with a common NDV genotype II vaccine (GII-vacc). To overcome this, the killed GVII vaccine has been used to prevent NDV outbreaks. However, the debate about vaccine differences remains ongoing. Hence, this study investigated the difference in chickens' responses to the two vaccines at the molecular level. The spleen transcriptomes from vaccinated chickens reveal that GVII-vacc affected the immune response by downregulating neuroinflammation. It also enhanced a synaptogenesis pathway that operates typically in the nervous system, suggesting a mechanism for the neurotrophic effect of this strain. We speculated that the down-regulated immune system regulation correlated with protecting the nervous system from excess leukocytes and cytokine activity. In contrast, GII-vacc inhibited apoptosis by downregulating PERK/ATF4/CHOP as part of the unfolded protein response pathway but did not affect the expression of the same synaptogenesis pathway. Thus, the application of GVII-vacc needs to be considered in countries where GVII is the leading cause of NDV outbreaks. The predicted molecular signatures may also be used in developing new vaccines that trigger specific genes in the immune system in combating NDV outbreaks.
Collapse
Affiliation(s)
- Putri Pandarangga
- Departemen Klinik, Reproduksi, dan Patologi, Fakultas Kedokteran dan Kedokteran Hewan, Universitas Nusa Cendana, Kupang 85001, Indonesia;
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
| | - Phuong Thi Kim Doan
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
- Department of Veterinary Medicine, Tay Nguyen University, Buon Ma Thuot 630000, Vietnam
| | - Rick Tearle
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (R.T.); (W.Y.L.); (Y.R.)
| | - Wai Yee Low
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (R.T.); (W.Y.L.); (Y.R.)
| | - Yan Ren
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (R.T.); (W.Y.L.); (Y.R.)
| | - Hanh Thi Hong Nguyen
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
| | | | - Farhid Hemmatzadeh
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide 5371, Australia; (P.T.K.D.); (H.T.H.N.)
| |
Collapse
|
498
|
Meier M, Becker S, Levine E, DuFresne O, Foster K, Moore J, Burnett FN, Hermanns VC, Heath SP, Abdelsaid M, Coucha M. Timing matters in the use of renin-angiotensin system modulators and COVID-related cognitive and cerebrovascular dysfunction. PLoS One 2024; 19:e0304135. [PMID: 39074114 DOI: 10.1371/journal.pone.0304135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 05/07/2024] [Indexed: 07/31/2024] Open
Abstract
Renin-angiotensin system (RAS) modulators, including Angiotensin receptor blockers (ARB) and angiotensin-converting enzyme inhibitors (ACEI), are effective medications for controlling blood pressure. Cognitive deficits, including lack of concentration, memory loss, and confusion, were reported after COVID-19 infection. ARBs or ACEI increase the expression of angiotensin-converting enzyme-2 (ACE-2), a functional receptor that allows binding of SARS-CoV-2 spike protein for cellular invasion. To date, the association between the use of RAS modulators and the severity of COVID-19 cognitive dysfunction is still controversial. PURPOSE This study addressed the following questions: 1) Does prior treatment with RAS modulator worsen COVID-19-induced cerebrovascular and cognitive dysfunction? 2) Can post-treatment with RAS modulator improve cognitive performance and cerebrovascular function following COVID-19? We hypothesize that pre-treatment exacerbates COVID-19-induced detrimental effects while post-treatment displays protective effects. METHODS Clinical study: Patients diagnosed with COVID-19 between May 2020 and December 2022 were identified through the electronic medical record system. Inclusion criteria comprised a documented medical history of hypertension treated with at least one antihypertensive medication. Subsequently, patients were categorized into two groups: those who had been prescribed ACEIs or ARBs before admission and those who had not received such treatment before admission. Each patient was evaluated on admission for signs of neurologic dysfunction. Pre-clinical study: Humanized ACE-2 transgenic knock-in mice received the SARS-CoV-2 spike protein via jugular vein injection for 2 weeks. One group had received Losartan (10 mg/kg), an ARB, in their drinking water for two weeks before the injection, while the other group began Losartan treatment after the spike protein injection. Cognitive functions, cerebral blood flow, and cerebrovascular density were determined in all experimental groups. Moreover, vascular inflammation and cell death were assessed. RESULTS Signs of neurological dysfunction were observed in 97 out of 177 patients (51%) taking ACEIs/ARBs prior to admission, compared to 32 out of 118 patients (27%) not receiving ACEI or ARBs. In animal studies, spike protein injection increased vascular inflammation, increased endothelial cell apoptosis, and reduced cerebrovascular density. In parallel, spike protein decreased cerebral blood flow and cognitive function. Our results showed that pretreatment with Losartan exacerbated these effects. However, post-treatment with Losartan prevented spike protein-induced vascular and neurological dysfunctions. CONCLUSION Our clinical data showed that the use of RAS modulators before encountering COVID-19 can initially exacerbate vascular and neurological dysfunctions. Similar findings were demonstrated in the in-vivo experiments; however, the protective effects of targeting the RAS become apparent in the animal model when the treatment is initiated after spike protein injection.
Collapse
Affiliation(s)
- Mackenzi Meier
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Sara Becker
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Erica Levine
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Oriana DuFresne
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Kaleigh Foster
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Joshua Moore
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Faith N Burnett
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Veronica C Hermanns
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Stan P Heath
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Mohammed Abdelsaid
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, Georgia, United States of America
| |
Collapse
|
499
|
de Souza AJS, de Souza AF, Zimpel CK, Ayupe MC, de Araújo MV, Machado RRG, Salles E, Salgado CL, Tavares MS, Silva-Pereira TT, de Souza PC, Durigon EL, Heinemann MB, Brandão PE, da Fonseca DM, Guimarães AMDS, de Sá LRM. Hepatic endotheliitis in Golden Syrian hamsters (Mesocricetus auratus) experimentally infected with SARS-CoV-2. Rev Inst Med Trop Sao Paulo 2024; 66:e44. [PMID: 39082483 PMCID: PMC11295288 DOI: 10.1590/s1678-9946202466044] [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: 03/05/2024] [Accepted: 06/03/2024] [Indexed: 08/04/2024] Open
Abstract
Hepatic injuries in COVID-19 are not yet fully understood and indirect pathways (without viral replication in the liver) have been associated with the activation of vascular mechanisms of liver injury in humans infected with SARS-CoV-2. Golden Syrian hamsters are an effective model for experimental reproduction of moderate and self-limiting lung disease during SARS-CoV-2 infection. As observed in humans, this experimental model reproduces lesions of bronchointerstitial pneumonia and pulmonary vascular lesions, including endotheliitis (attachment of lymphoid cells to the luminal surface of endothelium). Extrapulmonary vascular lesions are well documented in COVID-19, but such extrapulmonary vascular lesions have not yet been described in the Golden Syrian hamster model of SARS-CoV-2 infection. The study aimed to evaluate microscopic liver lesions in Golden Syrian hamsters experimentally infected with SARS-CoV-2. In total, 38 conventional Golden Syrian hamsters, divided into infected group (n=24) and mock-infected group (n=14), were euthanized at 2-, 3-, 4-, 5-, 7-, 14-, and 15-days post infection with SARS-CoV-2. Liver fragments were evaluated by histopathology and immunohistochemical detection of SARS-CoV-2 Spike S2 antigens. The frequencies of portal vein endotheliitis, lobular activity, hepatocellular degeneration, and lobular vascular changes were higher among SARS-CoV-2-infected animals. Spike S2 antigen was not detected in liver. The main results indicate that SARS-CoV-2 infection exacerbated vascular and inflammatory lesions in the liver of hamsters with pre-existing hepatitis of unknown origin. A potential application of this animal model in studies of the pathogenesis and evolution of liver lesions associated with SARS-CoV-2 infection still needs further evaluation.
Collapse
Affiliation(s)
- Alex Junior Souza de Souza
- Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Departamento de Patologia, São Paulo, São Paulo, Brazil
| | - Antônio Francisco de Souza
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
| | - Cristina Kraemer Zimpel
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
- Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Departamento de Medicina Veterinária Preventiva e Saúde Animal, São Paulo, São Paulo, Brazil
| | - Marina Caçador Ayupe
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Imunologia, São Paulo, São Paulo, Brazil
| | - Marcelo Valdemir de Araújo
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Imunologia, São Paulo, São Paulo, Brazil
- Instituto Butantan, Centro de Desenvolvimento e Inovação, Laboratório de Virologia, São Paulo, São Paulo, Brazil
| | - Rafael Rahal Guaragna Machado
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
| | - Erika Salles
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Imunologia, São Paulo, São Paulo, Brazil
| | - Caio Loureiro Salgado
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Imunologia, São Paulo, São Paulo, Brazil
| | - Mariana Silva Tavares
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
| | - Taiana Tainá Silva-Pereira
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
| | - Paula Carolina de Souza
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Imunologia, São Paulo, São Paulo, Brazil
| | - Edison Luiz Durigon
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
| | - Marcos Bryan Heinemann
- Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Departamento de Medicina Veterinária Preventiva e Saúde Animal, São Paulo, São Paulo, Brazil
| | - Paulo Eduardo Brandão
- Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Departamento de Medicina Veterinária Preventiva e Saúde Animal, São Paulo, São Paulo, Brazil
| | - Denise Morais da Fonseca
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Imunologia, São Paulo, São Paulo, Brazil
| | - Ana Marcia de Sá Guimarães
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, São Paulo, Brazil
| | - Lilian Rose Marques de Sá
- Universidade de São Paulo, Faculdade de Medicina Veterinária e Zootecnia, Departamento de Patologia, São Paulo, São Paulo, Brazil
| |
Collapse
|
500
|
Bouare N, Delwaide J. Interleukin-mediated therapies in liver diseases and comorbidity effects. World J Hepatol 2024; 16:980-989. [PMID: 39086534 PMCID: PMC11287617 DOI: 10.4254/wjh.v16.i7.980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 07/26/2024] Open
Abstract
Cytokines like interleukins (ILs) play important roles in inflammation and innate immune. Yang and Zhang carried out an interesting study related to ILs and hepatic diseases. They described the role of ILs in the pathogenesis and resolution of hepatic disorders. The authors summarized alcohol-related liver disease and virus-induced hepatitis, as far as clinical studies a fortiori carried out on IL-mediated treatments pertaining to these dysfunctions. This editorial contributes to the review by Yang and Zhang titled, "Interleukins in liver disease treatment", and focuses on therapies mediated by ILs in comorbid liver diseases. The documentary search was conducted on recent pertinent literature, primarily using the Google Scholar and PubMed databases.
Collapse
Affiliation(s)
- Nouhoum Bouare
- Department of Quality, Hygien, Biosafety/Biosecurity and Pharmacovigilence, National Institute of Public Health, Bamako 1771, Mali.
| | - Jean Delwaide
- Department of Gastroenterology and Hepatology, CHULiege, Liege 4000, Belgium
| |
Collapse
|