1
|
Taylor RA, Xiao S, Carias AM, McRaven MD, Thakkar DN, Araínga M, Lorenzo-Redondo R, Allen EJ, Rogers KA, Kumarapperuma SC, Gong S, Anderson MR, Thomas Y, Madden PJ, Corti D, Cameroni E, Lanzavecchia A, Goins B, Fox P, Villinger FJ, Ruprecht RM, Hope TJ. PET/CT Targeted Tissue Sampling Reveals Intravenously Administered HGN194 IgG1 Affects HIV Distribution after Rectal Exposure. AIDS Res Hum Retroviruses 2024; 40:637-648. [PMID: 39104250 DOI: 10.1089/aid.2024.0019] [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: 08/07/2024] Open
Abstract
Neutralizing monoclonal antibodies hold great potential for prevention of human immunodeficiency virus (HIV) acquisition. IgG is the most abundant antibody in human serum, has a long half-life, and potent effector functions, making it a prime candidate for an HIV prevention therapeutic. We combined Positron Emission Tomography imaging and fluorescent microscopy of 64Cu-labeled, photoactivatable-green fluorescent protein HIV (PA-GFP-BaL) and fluorescently labeled HGN194 IgG1 to determine whether intravenously instilled IgG influences viral interaction with mucosal barriers and viral penetration in colorectal tissue 2 h after rectal viral challenge. Our results show that IgG1 did not alter the number of virions found throughout the colon or viral penetration into the epithelium of the rectum or descending colon. A minor increase in virions was observed in the transverse colon of IgG1 treated animals. Overall, the number of viral particles found in the mesenteric lymph nodes was low. However, IgG1 administration resulted in a significant reduction of virions found in mesenteric lymph nodes. Taken together, our results show that HGN194 IgG1 does not prevent virions from penetrating into the colorectal mucosa but may perturb HIV virion access to the lymphatic system.
Collapse
Affiliation(s)
- Roslyn A Taylor
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sixia Xiao
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ann M Carias
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael D McRaven
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Divya N Thakkar
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mariluz Araínga
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Ramon Lorenzo-Redondo
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Institute for Global Health, Chicago, Illinois, USA
| | - Edward J Allen
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kenneth A Rogers
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Sidath C Kumarapperuma
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Siqi Gong
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, USA
- Texas Biomedical Research Institute and Southwest National Primate Research Center, San Antonio, Texas, USA
| | - Meegan R Anderson
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yanique Thomas
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Patrick J Madden
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Davide Corti
- Humabs Biomed, a subsidiary of Vir Biotechnology Inc, Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs Biomed, a subsidiary of Vir Biotechnology Inc, Bellinzona, Switzerland
| | | | - Beth Goins
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Peter Fox
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Francois J Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Ruth M Ruprecht
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, USA
- Texas Biomedical Research Institute and Southwest National Primate Research Center, San Antonio, Texas, USA
| | - Thomas J Hope
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
2
|
Asediya VS, Anjaria PA, Mathakiya RA, Koringa PG, Nayak JB, Bisht D, Fulmali D, Patel VA, Desai DN. Vaccine development using artificial intelligence and machine learning: A review. Int J Biol Macromol 2024; 282:136643. [PMID: 39426778 DOI: 10.1016/j.ijbiomac.2024.136643] [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: 06/01/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The COVID-19 pandemic has underscored the critical importance of effective vaccines, yet their development is a challenging and demanding process. It requires identifying antigens that elicit protective immunity, selecting adjuvants that enhance immunogenicity, and designing delivery systems that ensure optimal efficacy. Artificial intelligence (AI) can facilitate this process by using machine learning methods to analyze large and diverse datasets, suggest novel vaccine candidates, and refine their design and predict their performance. This review explores how AI can be applied to various aspects of vaccine development, such as predicting immune response from protein sequences, discovering adjuvants, optimizing vaccine doses, modeling vaccine supply chains, and predicting protein structures. We also address the challenges and ethical issues that emerge from the use of AI in vaccine development, such as data privacy, algorithmic bias, and health data sensitivity. We contend that AI has immense potential to accelerate vaccine development and respond to future pandemics, but it also requires careful attention to the quality and validity of the data and methods used.
Collapse
Affiliation(s)
| | | | | | | | | | - Deepanker Bisht
- Indian Veterinary Research Institute, Izatnagar, U.P., India
| | | | | | | |
Collapse
|
3
|
Sigawi T, Israeli A, Ilan Y. Harnessing Variability Signatures and Biological Noise May Enhance Immunotherapies' Efficacy and Act as Novel Biomarkers for Diagnosing and Monitoring Immune-Associated Disorders. Immunotargets Ther 2024; 13:525-539. [PMID: 39431244 PMCID: PMC11488351 DOI: 10.2147/itt.s477841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
Lack of response to immunotherapies poses a significant challenge in treating immune-mediated disorders and cancers. While the mechanisms associated with poor responsiveness are not well defined and change between and among subjects, the current methods for overcoming the loss of response are insufficient. The Constrained Disorder Principle (CDP) explains biological systems based on their inherent variability, bounded by dynamic boundaries that change in response to internal and external perturbations. Inter and intra-subject variability characterize the immune system, making it difficult to provide a single therapeutic regimen to all patients and even the same patients over time. The dynamicity of the immune variability is also a significant challenge for personalizing immunotherapies. The CDP-based second-generation artificial intelligence system is an outcome-based dynamic platform that incorporates personalized variability signatures into the therapeutic regimen and may provide methods for improving the response and overcoming the loss of response to treatments. The signatures of immune variability may also offer a method for identifying new biomarkers for early diagnosis, monitoring immune-related disorders, and evaluating the response to treatments.
Collapse
Affiliation(s)
- Tal Sigawi
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Adir Israeli
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Yaron Ilan
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
4
|
Chen C, Su Y, Li S, Man C, Jiang Y, Qu B, Yang X, Guo L. Advances in oligosaccharides and polysaccharides with different structures as wall materials for probiotics delivery: A review. Int J Biol Macromol 2024; 277:134468. [PMID: 39217037 DOI: 10.1016/j.ijbiomac.2024.134468] [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: 03/31/2024] [Revised: 06/29/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Probiotics are active microorganisms that are beneficial to the health of the host. However, probiotics are highly sensitive to the external environment, and are susceptible to a variety of factors that reduce their activity during production, storage, and use. Microencapsulation is an effective method that enhances probiotic activity. Macromolecules like polysaccharides, who classified as biologically active prebiotics, have attracted significant attention for their utility in probiotic microencapsulation. This article summarized the types of commonly used microencapsulation materials and their structural characteristics from the perspective of polysaccharides prebiotics. It also discussed recent advancements, probiotic-prebiotic microcapsule-based modulation of the immune system, as well as the associated limitations. Furthermore, the advantages and disadvantages of eight prebiotics as microencapsulation wall materials. The honeycomb structure of β-glucan enhances the bioavailability of probiotics, while, fructooligosaccharide and galactooligosaccharides improve microbead structure to tightly encapsulate probiotics. The terminal reducing groups of isomaltooligosaccharides and the free hydroxyl groups in xylooligosaccharides also positively affect the structure of microcapsules. Prebiotics not only enhance the survival rate and biological activity of probiotics as embedding materials during storage, but also exert their own probiotic effects. Collectively, prebiotics holds great promise as microencapsulation materials for probiotics delivery.
Collapse
Affiliation(s)
- Chen Chen
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yue Su
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Shihang Li
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Bo Qu
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyan Yang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Ling Guo
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
5
|
Alzahrani AR, Mohamed DI, Abo Nahas HH, Alaa El-Din Aly El-Waseef D, Altamimi AS, Youssef IH, Ibrahim IAA, Mohamed SMY, Sabry YG, Falemban AH, Elhawary NA, Bamagous GA, Jaremko M, Saied EM. Trimetazidine Alleviates Bleomycin-Induced Pulmonary Fibrosis by Targeting the Long Noncoding RNA CBR3-AS1-Mediated miRNA-29 and Resistin-Like Molecule alpha 1: Deciphering a Novel Trifecta Role of LncRNA CBR3-AS1/miRNA-29/FIZZ1 Axis in Lung Fibrosis. Drug Des Devel Ther 2024; 18:3959-3986. [PMID: 39252766 PMCID: PMC11382803 DOI: 10.2147/dddt.s463626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction Pulmonary fibrosis (PF) and tissue remodeling can greatly impair pulmonary function and often lead to fatal outcomes. Methodology In the present study, we explored a novel molecular interplay of long noncoding (Lnc) RNA CBR3-AS1/ miRNA-29/ FIZZ1 axis in moderating the inflammatory processes, immunological responses, and oxidative stress pathways in bleomycin (BLM)-induced lung fibrosis. Furthermore, we investigated the pharmacological potential of Trimetazidine (TMZ) in ameliorating lung fibrosis. Results Our results revealed that the BLM-treated group exhibited a significant upregulation in the expression of epigenetic regulators, lncRNA CBR3-AS1 and FIZZ1, compared to the control group (P<0.0001), along with the downregulation of miRNA-29 expression. Furthermore, Correlation analysis showed a significant positive association between lnc CBR3-AS1 and FIZZ1 (R=0.7723, p<0.05) and a significant negative association between miRNA-29 and FIZZ1 (R=-0.7535, p<0.05), suggesting lnc CBR3-AS1 as an epigenetic regulator of FIZZ1 in lung fibrosis. BLM treatment significantly increased the expression of Notch, Jagged1, Smad3, TGFB1, and hydroxyproline. Interestingly, the administration of TMZ demonstrated the ability to attenuate the deterioration effects caused by BLM treatment, as indicated by biochemical and histological analyses. Our investigations revealed that the therapeutic potential of TMZ as an antifibrotic drug could be ascribed to its ability to directly target the epigenetic regulators lncRNA CBR3-AS1/ miRNA-29/ FIZZ1, which in turn resulted in the mitigation of lung fibrosis. Histological and immunohistochemical analyses further validated the potential antifibrotic effects of TMZ by mitigating the structural damage associated with fibrosis. Discussion Taken together, our study showed for the first time the interplay between epigenetic lncRNAs CBR3-AS1 and miRNA-29 in lung fibrosis and demonstrated that FIZZ1 could be a downregulatory gene for lncRNA CBR3-AS1 and miRNA-29. Our key findings demonstrate that TMZ significantly reduces the expression of fibrotic, oxidative stress, immunomodulatory, and inflammatory markers, along with epigenetic regulators associated with lung fibrosis. This validates its potential as an effective antifibrotic agent by targeting the CBR3-AS1/miRNA-29/FIZZ1 axis.
Collapse
Affiliation(s)
- Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Doaa I Mohamed
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Abdulmalik S Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Ibrahim H Youssef
- Department of Chest Diseases, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Soha M Y Mohamed
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yasmine Gamal Sabry
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Alaa H Falemban
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Nasser Attia Elhawary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Ghazi A Bamagous
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative and Red Sea Research Center, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Essa M Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
- Institute for Chemistry, Humboldt Universität Zu Berlin, Berlin, Germany
| |
Collapse
|
6
|
Tan J, Wang D, Dong W, Nian L, Zhang F, Zhao H, Zhang J, Feng Y. Comprehensive Analysis of CCAAT/Enhancer Binding Protein Family in Ovarian Cancer. Cancer Inform 2024; 23:11769351241275877. [PMID: 39238655 PMCID: PMC11375656 DOI: 10.1177/11769351241275877] [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: 05/02/2024] [Accepted: 07/28/2024] [Indexed: 09/07/2024] Open
Abstract
Background Ovarian cancer has brought serious threats to female health. CCAAT/enhancer binding proteins (C/EBPs) are key transcription factors involved in ovarian cancer. Therefore, comprehensive profiling C/EBPs in ovarian cancer is needed. Methods A comprehensive analysis concerning C/EBPs in ovarian cancer was performed. Firstly, detailed expression of C/EBP family members was integrally retrieved and then confirmed using immunohistochemistry. The regulatory effects and transcription regulatory functions of C/EBPs were studied by using regulatory network analysis and enrichment analysis. Using survival analysis, receiver operating characteristic curve analysis, and target-disease association analysis, the predictive prognostic value of C/EBPs on survival and drug responsiveness was systematically evaluated. The effects of C/EBPs on tumor immune infiltration were also assessed. Results Ovarian cancer tissues expressed increased CEBPA, CEBPB, and CEBPG but decreased CEBPD when compared with normal control tissues. The overall alteration frequency of C/EBPs in ovarian cancer was approaching 30%. C/EBP family members formed a reciprocal regulatory network involving carcinogenesis and had pivotal transcription regulatory functions. C/EBPs could affect survival of ovarian cancer and correlated with poor survival outcomes (OS: HR = 1.40, P = .0053 and PFS: HR = 1.41, P = .0036). Besides, expression of CEBPA, CEBPB, CEBPD, and CEBPE could predict platinum and taxane responsiveness of ovarian cancer. C/EBPs also affected immune infiltration of ovarian cancer. Conclusions C/EBPs were closely involved in ovarian cancer and exerted multiple biological functions. C/EBPs could be exploited as prognostic and predictive biomarkers in ovarian cancer.
Collapse
Affiliation(s)
- Jiahong Tan
- Department of Obstetrics and Gynecology, National Key Clinical Specialty of Gynecology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Daoqi Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Wei Dong
- Department of Obstetrics and Gynecology, National Key Clinical Specialty of Gynecology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Lei Nian
- Department of Obstetrics and Gynecology, National Key Clinical Specialty of Gynecology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Fen Zhang
- Department of Obstetrics and Gynecology, National Key Clinical Specialty of Gynecology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Han Zhao
- Department of Obstetrics and Gynecology, National Key Clinical Specialty of Gynecology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Jie Zhang
- Department of Obstetrics and Gynecology, National Key Clinical Specialty of Gynecology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Yun Feng
- Department of Obstetrics and Gynecology, National Key Clinical Specialty of Gynecology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, People's Republic of China
| |
Collapse
|
7
|
Muñoz-Melero M, Biswas M. Role of FoxP3 + Regulatory T Cells in Modulating Immune Responses to Adeno-Associated Virus Gene Therapy. Hum Gene Ther 2024; 35:439-450. [PMID: 38450566 PMCID: PMC11302314 DOI: 10.1089/hum.2023.227] [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: 12/27/2023] [Accepted: 03/04/2024] [Indexed: 03/08/2024] Open
Abstract
Adeno-associated virus (AAV) gene therapy is making rapid strides owing to its wide range of therapeutic applications. However, development of serious immune responses to the capsid antigen or the therapeutic transgene product hinders its full clinical impact. Immune suppressive (IS) drug treatments have been used in various clinical trials to prevent the deleterious effects of cytotoxic T cells to the viral vector or transgene, although there is no consensus on the best treatment regimen, dosage, or schedule. Regulatory T cells (Tregs) are crucial for maintaining tolerance against self or nonself antigens. Of importance, Tregs also play an important role in dampening immune responses to AAV gene therapy, including tolerance induction to the transgene product. Approaches to harness the tolerogenic effect of Tregs include the use of selective IS drugs that expand existing Tregs, and skew activated conventional T cells into antigen-specific peripherally induced Tregs. In addition, Tregs can be expanded ex vivo and delivered as cellular therapy. Furthermore, receptor engineering can be used to increase the potency and specificity of Tregs allowing for suppression at lower doses and reducing the risk of disrupting protective immunity. Because immune-mediated toxicities to AAV vectors are a concern in the clinic, strategies that can enhance or preserve Treg function should be considered to improve both the safety and efficacy of AAV gene therapy.
Collapse
Affiliation(s)
- Maite Muñoz-Melero
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
8
|
Sangiorgio G, Calvo M, Migliorisi G, Campanile F, Stefani S. The Impact of Enterococcus spp. in the Immunocompromised Host: A Comprehensive Review. Pathogens 2024; 13:409. [PMID: 38787261 PMCID: PMC11124283 DOI: 10.3390/pathogens13050409] [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: 04/20/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
The immunocompromised host is usually vulnerable to infectious diseases due to broad-spectrum treatments and immunological dysregulation. The Enterococcus genus consists of normal gut commensals, which acquire a leading role in infective processes among individuals with compromised immune systems. These microorganisms may express a potential virulence and resistance spectrum, enabling their function as severe pathogens. The Enterococcus spp. infections in immunocompromised hosts appear to be difficult to resolve due to the immunological response impairment and the possibility of facing antimicrobial-resistant strains. As regards the related risk factors, several data demonstrated that prior antibiotic exposure, medical device insertion, prolonged hospitalization and surgical interventions may lead to Enterococcus overgrowth, antibiotic resistance and spread among critical healthcare settings. Herein, we present a comprehensive review of Enterococcus spp. in the immunocompromised host, summarizing the available knowledge about virulence factors, antimicrobial-resistance mechanisms and host-pathogen interaction. The review ultimately yearns for more substantial support to further investigations about enterococcal infections and immunocompromised host response.
Collapse
Affiliation(s)
- Giuseppe Sangiorgio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (F.C.); (S.S.)
| | - Maddalena Calvo
- U.O.C. Laboratory Analysis Unit, University Hospital Policlinico-San Marco, Via Santa Sofia 78, 95123 Catania, Italy; (M.C.); (G.M.)
| | - Giuseppe Migliorisi
- U.O.C. Laboratory Analysis Unit, University Hospital Policlinico-San Marco, Via Santa Sofia 78, 95123 Catania, Italy; (M.C.); (G.M.)
| | - Floriana Campanile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (F.C.); (S.S.)
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; (F.C.); (S.S.)
- U.O.C. Laboratory Analysis Unit, University Hospital Policlinico-San Marco, Via Santa Sofia 78, 95123 Catania, Italy; (M.C.); (G.M.)
| |
Collapse
|
9
|
Sira EMJS, Banico EC, Odchimar NMO, Fajardo LE, Fremista FF, Refuerzo HAB, Dictado APA, Orosco FL. Immunoinformatics approach for designing a multiepitope subunit vaccine against porcine epidemic diarrhea virus genotype IIA spike protein. Open Vet J 2024; 14:1224-1242. [PMID: 38938443 PMCID: PMC11199741 DOI: 10.5455/ovj.2024.v14.i5.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/26/2024] [Indexed: 06/29/2024] Open
Abstract
Background Porcine epidemic diarrhea (PED), caused by the porcine epidemic diarrhea virus (PEDV), is associated with high mortality and morbidity rates, especially in neonatal pigs. This has resulted in significant economic losses for the pig industry. PEDV genotype II-based vaccines were found to confer better immunity against both heterologous and homologous challenges; specifically, spike (S) proteins, which are known to play a significant role during infection, are ideal for vaccine development. Aim This study aims to design a multi-epitope subunit vaccine targeting the S protein of the PEDV GIIa strain using an immunoinformatics approach. Methods Various bioinformatics tools were used to predict HTL, CTL, and B-cell epitopes. The epitopes were connected using appropriate linkers and conjugated with the CTB adjuvant and M-ligand. The final multiepitope vaccine construct (fMEVc) was then docked to toll-like receptor 4 (TLR4). The stability of the fMEVc-TLR4 complex was then simulated using GROMACS. C-immsim was then used to predict the in vitro immune response of the fMEVc. Results Six epitopes were predicted to induce antibody production, ten epitopes were predicted to induce CTL responses, and four epitopes were predicted to induce HTL responses. The assembled epitopes conjugated with the CTB adjuvant and M-ligand, fMEVc, is antigenic, non-allergenic, stable, and soluble. The construct showed a favorable binding affinity for TLR4, and the protein complex was shown to be stable through molecular dynamics simulations. A robust immune response was induced after immunization, as demonstrated through immune stimulation. Conclusion In conclusion, the multi-epitope subunit vaccine construct for PEDV designed in this study exhibits promising antigenicity, stability, and immunogenicity, eliciting robust immune responses and suggesting its potential as a candidate for further vaccine development.
Collapse
Affiliation(s)
- Ella Mae Joy S. Sira
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Edward C. Banico
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Nyzar Mabeth O. Odchimar
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Lauren Emily Fajardo
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
| | - Ferdinand F. Fremista
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Philippines
| | | | - Ana Patrisha A. Dictado
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Philippines
| | - Fredmoore L. Orosco
- Virology and Vaccine Research and Development Program, Industrial Technology Development Institute, Department of Science and Technology, Taguig City, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Philippines
- S&T Fellows Program, Department of Science and Technology, Taguig City, Philippines
| |
Collapse
|
10
|
Li J, Ding J, Wu H, Lu C, Wu J, Luo Q. Tat-CIRP Peptide Facilitates Frozen Wound Healing by Ameliorating Inflammation and Promoting Angiogenesis. J Inflamm Res 2024; 17:2205-2215. [PMID: 38623470 PMCID: PMC11017987 DOI: 10.2147/jir.s450288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/04/2024] [Indexed: 04/17/2024] Open
Abstract
Background Frostbite is a chemia resulting from cold-induced skin damage. The process of frostbite is often accompanied by inflammation, and the therapeutic strategies focusing on anti-inflammation are the main direction to data. Tat-CIRP is a 15 amino acid peptide containing HIV protein and cold-inducible RNA-binding protein (CIRP), which is believed to compete with endogenous CIRP for myeloid differentiation 2 (MD2) binding. This study aims to investigate the efficacy of Tat-CIRP in the treatment of frostbite. Methods A mouse model of frostbite was established, and on the first day after frostbite occurrence, Tat-CIRP peptide was administered intravenously via the tail with a dosage interval of one day for a total of three doses. Frozen mouse skin sections were subjected to histological analysis, including hematoxylin-eosin (HE) staining, Masson staining, and immunohistochemical examination. Western blotting was performed to detect the expression level of Ki-67 in mouse skin tissue. Results One day after frostbite, mice exhibited skin swelling and a solid appearance. From day 1 to 5 after frostbite, MD2 expression was significantly upregulated, while CIRP expression was downregulated. Compared to the frostbite group, mice treated with Tat-CIRP showed accelerated frostbite recovery, reduced levels of inflammatory factors and MD2. Furthermore, the expression of cell proliferation-associated protein Ki-67 and angiogenesis-related protein CD31 was upregulated. Conclusion Tat-CIRP promotes frozen wound healing via inhibiting inflammation and promoting angiogenesis in frostbitten mice.
Collapse
Affiliation(s)
- Jiayan Li
- Department of Hypoxic Biomedicine, Institute of Special Environmental Medicine and Coinnovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People’s Republic of China
| | - Jie Ding
- Department of Hypoxic Biomedicine, Institute of Special Environmental Medicine and Coinnovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People’s Republic of China
| | - Haoyang Wu
- Department of Hypoxic Biomedicine, Institute of Special Environmental Medicine and Coinnovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People’s Republic of China
| | - Chenyan Lu
- Department of Hypoxic Biomedicine, Institute of Special Environmental Medicine and Coinnovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People’s Republic of China
| | - Jian Wu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| | - Qianqian Luo
- Department of Hypoxic Biomedicine, Institute of Special Environmental Medicine and Coinnovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People’s Republic of China
| |
Collapse
|
11
|
Zhao Y, Wang H, Wu Z, Zhu Y, Wang J. Case Report: A Rare Case of Iodixanol-Induced Anaphylactic Shock in Cerebral Angiography. J Asthma Allergy 2024; 17:361-367. [PMID: 38623449 PMCID: PMC11018123 DOI: 10.2147/jaa.s460263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
Background Adverse reactions induced by isoosmolar contrast medium (iodixanol) are mostly mild, with rashes and headaches being the most common. Although anaphylactic shock has been reported, no related incidents have been documented on cerebral angiography. Objective This article reports a serious case of anaphylactic shock possibly induced by iodixanol and provides an overview of the case report. Case Summary A 65-year-old female with persistent headaches for nearly six months and CTA examination revealed multiple intracranial aneurysms. After two treatments, she returned to the hospital for aneurysm of reexamination a month ago. Following a preoperative assessment, cerebral angiography was performed. Three minutes after the procedure, the patient experienced dizziness, increased heart rate, followed by hypotension (BP 90/43 mm Hg), a sudden drop-in heart rate (HR 68 bpm), and a drop in SpO2 to 92%. Intravenous dexamethasone for anti-allergic were administered immediately, along with therapy through oxygen-inhalation. However, the patient then developed limb convulsions, unresponsiveness, and was urgently given diazepam for sedation and sputum aspiration to maintain airway patency. Blood pressure decrease to 53/29 mm Hg, and SpO2 readings were unavailable. Intravenous dopamine to elevates blood pressure, and assists breathing by intubating in the endotracheal. After 3 minutes, as the blood pressure remained undetectable, intermittent intravenous epinephrine 1mg was administered to raise the blood pressure, gradually restoring it to 126/90 mm Hg, and SpO2 increased to 95%. The patient was diagnosed with iodixanol-induced anaphylactic shock and urgently transferred to the NICU for monitoring and treatment. The patient died despite immediate treatment. Conclusion A 65-year-old female developed serious anaphylactic shock during cerebral angiography after receiving iodixanol. Although iodixanol is considered one of the safest iodinated contrast mediums (ICM), clinicians should be aware of its the potential for serious hypersensitivity reactions that can lead to fatal and life-threatening events.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| | - Hua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Zhengjun Wu
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| | - Yunxiang Zhu
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, People’s Republic of China
| |
Collapse
|
12
|
Feng L, Li J, Qian Z, Li C, Gao D, Wang Y, Xie W, Cai Y, Tong Z, Liang L. Comprehensive Nomograms Using Routine Biomarkers Beyond Eosinophil Levels: Enhancing Predictability of Corticosteroid Treatment Outcomes in AECOPD. J Inflamm Res 2024; 17:1511-1526. [PMID: 38476472 PMCID: PMC10929658 DOI: 10.2147/jir.s450447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose Patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD) exhibit heterogeneous responses to corticosteroid treatment. We aimed to determine whether combining eosinophil levels with other routine clinical indicators can enhance the predictability of corticosteroid treatment outcomes and to come up with a scoring system. Patients and Methods Consecutive patients admitted with AECOPD receiving corticosteroid treatment between July 2013 and March 2022 at Beijing Chao-Yang Hospital were retrospectively analyzed. Data on patients' demographics, smoking status, hospitalization for AECOPD in the previous year, comorbidities, blood laboratory tests, in-hospital treatment and clinical outcomes were collected. Least absolute shrinkage and selection operator (LASSO) regression and backward logistic regression were used for predictor selection, and predictive nomograms were developed. The discrimination and calibration of the nomograms were assessed using the area under the receiver operating curve (AUC) and calibration plots. Internal validation was performed using the 500-bootstrap method, and clinical utility was evaluated using decision curve analysis (DCA). Results Among the 3254 patients included, 804 (24.7%) had treatment failure. A nomogram of eosinophils, platelets, C-reactive protein (CRP), low density lipoprotein cholesterol, prognostic nutritional index (PNI), hospitalization for AECOPD in the previous year, ischemic heart diseases and chronic hepatic disease was developed to predict treatment failure for patients with a smoking history. For patients without a smoking history, a nomogram of CRP, PNI, ischemic heart diseases and chronic hepatic disease was developed. Although the AUCs of these two nomograms were only 0.644 and 0.647 respectively, they were significantly superior to predictions based solely on blood eosinophil levels. Conclusion We developed easy-to-use comprehensive nomograms utilizing readily available clinical biomarkers related to inflammation, nutrition and immunity, offering modestly enhanced predictive value for treatment outcomes in corticosteroid-treated patients with AECOPD. Further investigations into novel biomarkers and additional patient data are imperative to optimize the predictive performance.
Collapse
Affiliation(s)
- Lin Feng
- Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jiachen Li
- Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhenbei Qian
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Chenglong Li
- Heart and Vascular Health Research Center, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, People’s Republic of China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, People’s Republic of China
| | - Darui Gao
- Heart and Vascular Health Research Center, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, People’s Republic of China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, People’s Republic of China
| | - Yongqian Wang
- Heart and Vascular Health Research Center, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, People’s Republic of China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, People’s Republic of China
| | - Wuxiang Xie
- Heart and Vascular Health Research Center, Peking University Clinical Research Institute, Peking University First Hospital, Beijing, People’s Republic of China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, People’s Republic of China
| | - Yutong Cai
- Centre for Environmental Health and Sustainability, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Lirong Liang
- Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
13
|
Bai BYH, Reppell M, Smaoui N, Waring JF, Pivorunas V, Guay H, Lin S, Chanchlani N, Bewshea C, Goodhand JR, Kennedy NA, Ahmad T, Anderson CA. Baseline Expression of Immune Gene Modules in Blood is Associated With Primary Response to Anti-TNF Therapy in Crohn's Disease Patients. J Crohns Colitis 2024; 18:431-445. [PMID: 37776235 PMCID: PMC10906954 DOI: 10.1093/ecco-jcc/jjad166] [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: 06/06/2023] [Revised: 08/22/2023] [Accepted: 09/28/2023] [Indexed: 10/02/2023]
Abstract
BACKGROUND AND AIMS Anti-tumour necrosis factor [anti-TNF] therapy is widely used for the treatment of inflammatory bowel disease, yet many patients are primary non-responders, failing to respond to induction therapy. We aimed to identify blood gene expression differences between primary responders and primary non-responders to anti-TNF monoclonal antibodies [infliximab and adalimumab], and to predict response status from blood gene expression and clinical data. METHODS The Personalised Anti-TNF Therapy in Crohn's Disease [PANTS] study is a UK-wide prospective observational cohort study of anti-TNF therapy outcome in anti-TNF-naive Crohn's disease patients [ClinicalTrials.gov identifier: NCT03088449]. Blood gene expression in 324 unique patients was measured by RNA-sequencing at baseline [week 0], and at weeks 14, 30, and 54 after treatment initiation [total sample size = 814]. RESULTS After adjusting for clinical covariates and estimated blood cell composition, baseline expression of major histocompatibility complex, antigen presentation, myeloid cell enriched receptor, and other innate immune gene modules was significantly higher in anti-TNF responders vs non-responders. Expression changes from baseline to week 14 were generally of consistent direction but greater magnitude [i.e. amplified] in responders, but interferon-related genes were upregulated uniquely in non-responders. Expression differences between responders and non-responders observed at week 14 were maintained at weeks 30 and 54. Prediction of response status from baseline clinical data, cell composition, and module expression was poor. CONCLUSIONS Baseline gene module expression was associated with primary response to anti-TNF therapy in PANTS patients. However, these baseline expression differences did not predict response with sufficient sensitivity for clinical use.
Collapse
Affiliation(s)
- Benjamin Y H Bai
- Genomics of Inflammation and Immunity Group, Wellcome Sanger Institute, Hinxton, UK
- Postgraduate School of Life Sciences, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | - Simeng Lin
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, UK
| | - Neil Chanchlani
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, UK
| | - Claire Bewshea
- Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, UK
| | - James R Goodhand
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, UK
| | - Nicholas A Kennedy
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, UK
| | - Tariq Ahmad
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, UK
| | - Carl A Anderson
- Genomics of Inflammation and Immunity Group, Wellcome Sanger Institute, Hinxton, UK
| |
Collapse
|
14
|
Phillips A. Self-assembling DNA recognizes patterns. Nature 2024; 625:454-455. [PMID: 38233615 DOI: 10.1038/d41586-023-03997-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
|
15
|
Kalogeropoulos D, Kanavaros P, Vartholomatos G, Moussa G, Kalogeropoulos C. Cytokines in Immune-mediated "Non-infectious" Uveitis. Klin Monbl Augenheilkd 2023. [PMID: 38134911 DOI: 10.1055/a-2202-8704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Uveitis is a significant cause of ocular morbidity and accounts for approximately 5 - 10% of visual impairments worldwide, particularly among the working-age population. Infections are the cause of ~ 50% cases of uveitis, but it has been suggested that infection might also be implicated in the pathogenesis of immune-mediated "non-infectious" uveitis. There is growing evidence that cytokines (i.e., interleukins, interferons, etc.) are key mediators of immune-mediated "non-infectious" uveitis. For example, activation of the interleukin-23/interleukin-17 signalling pathway is involved in immune-mediated "non-infectious" uveitis. Studies in animal models have been important in investigating the role of cytokines in uveitis. Recent studies of clinical samples from patients with uveitis have allowed the measurement of a considerable array of cytokines even from very small sample volumes (e.g., aqueous and vitreous humour). The identification of complex patterns of cytokines may contribute to a better understanding of their potential pathogenetic role in uveitis as well as to an improved diagnostic and therapeutic approach to treat these potentially blinding pathologies. This review provides further insights into the putative pathobiological role of cytokines in immune-mediated "non-infectious" uveitis.
Collapse
Affiliation(s)
| | - Panagiotis Kanavaros
- Anatomy-Histology-Embryology, University of Ioannina, Faculty of Medicine, Greece
| | - Georgios Vartholomatos
- Hematology Laboratory, Unit of Molecular Biology, University General Hospital of Ioannina, Greece
| | - George Moussa
- Ophthalmology, Birmingham and Midland Eye Centre, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | | |
Collapse
|
16
|
Hasubek AL, Wang X, Zhang E, Kobus M, Chen J, Vandergrift LA, Kurreck A, Ehret F, Dinges S, Hohm A, Tilgner M, Buko A, Habbel P, Nowak J, Mercaldo ND, Gusev A, Feldman AS, Cheng LL. Differentiation of patients with and without prostate cancer using urine 1 H NMR metabolomics. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2023; 61:740-747. [PMID: 37654196 DOI: 10.1002/mrc.5391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/09/2023] [Accepted: 08/12/2023] [Indexed: 09/02/2023]
Abstract
Prostate cancer (PCa) is one of the most prevalent cancers in men worldwide. For its detection, serum prostate-specific antigen (PSA) screening is commonly used, despite its lack of specificity, high false positive rate, and inability to discriminate indolent from aggressive PCa. Following increases in serum PSA levels, clinicians often conduct prostate biopsies with or without advanced imaging. Nuclear magnetic resonance (NMR)-based metabolomics has proven to be promising for advancing early-detection and elucidation of disease progression, through the discovery and characterization of novel biomarkers. This retrospective study of urine-NMR samples, from prostate biopsy patients with and without PCa, identified several metabolites involved in energy metabolism, amino acid metabolism, and the hippuric acid pathway. Of note, lactate and hippurate-key metabolites involved in cellular proliferation and microbiome effects, respectively-were significantly altered, unveiling widespread metabolomic modifications associated with PCa development. These findings support urine metabolomics profiling as a promising strategy to identify new clinical biomarkers for PCa detection and diagnosis.
Collapse
Affiliation(s)
- Anna-Laura Hasubek
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoyu Wang
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ella Zhang
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marta Kobus
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jiashang Chen
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lindsey A Vandergrift
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Annika Kurreck
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Felix Ehret
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah Dinges
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Annika Hohm
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marlon Tilgner
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander Buko
- Human Metabolome Technologies, Boston, Massachusetts, USA
| | - Piet Habbel
- Charite - Universitatsmedizin Berlin, Berlin, Germany
| | - Johannes Nowak
- SRH Poliklinik Gera GmbH, Radiology Gotha, Gotha, Germany
- SRH University of Applied Health Sciences, Gera, Germany
| | - Nathaniel D Mercaldo
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew Gusev
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Adam S Feldman
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Leo L Cheng
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Dupont D, Robert M, Brenier-Pinchart M, Lefevre A, Wallon M, Pelloux H. Toxoplasma gondii, a plea for a thorough investigation of its oncogenic potential. Heliyon 2023; 9:e22147. [PMID: 38034818 PMCID: PMC10685377 DOI: 10.1016/j.heliyon.2023.e22147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/20/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
It is estimated that 30 % of the world's population harbours the parasite Toxoplasma gondii, particularly in the brain. Beyond its implication in potentially severe opportunistic or congenital infections, this persistence has long been considered as without consequence. However, certain data in animals and humans suggest that this carriage may be linked to various neuropsychiatric or neurodegenerative disorders. The hypothesis of a potential cerebral oncogenicity of the parasite is also emerging. In this personal view, we will present the epidemiological arguments in favour of an association between toxoplasmosis and cerebral malignancy, before considering the points that could underlie a potential causal link. More specifically, we will focus on the brain as the preferred location for T. gondii persistence and the propensity of this parasite to interfere with the apoptosis and cell cycle signalling pathways of their host cell.
Collapse
Affiliation(s)
- D. Dupont
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
- Physiologie intégrée du système d’éveil, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Faculté de Médecine, Université Claude Bernard Lyon 1, Bron, 69500, France
| | - M.G. Robert
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| | - M.P. Brenier-Pinchart
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| | - A. Lefevre
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
| | - M. Wallon
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
- Physiologie intégrée du système d’éveil, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Faculté de Médecine, Université Claude Bernard Lyon 1, Bron, 69500, France
| | - H. Pelloux
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| |
Collapse
|
18
|
Jacho D, Babaniamansour P, Osorio R, Toledano M, Rabino A, Garcia-Mata R, Yildirim-Ayan E. Deciphering the Cell-Specific Effect of Osteoblast-Macrophage Crosstalk in Periodontitis. Tissue Eng Part A 2023; 29:579-593. [PMID: 37639358 PMCID: PMC10659017 DOI: 10.1089/ten.tea.2023.0104] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
In periodontitis, the bone remodeling process is disrupted by the prevalent involvement of bacteria-induced proinflammatory macrophage cells and their interaction with osteoblast cells residing within the infected bone tissue. The complex interaction between the cells needs to be deciphered to understand the dominant player in tipping the balance from osteogenesis to osteoclastogenesis. Yet, only a few studies have examined the crosstalk interaction between osteoblasts and macrophages using biomimetic three-dimensional (3D) tissue-like matrices. In this study, we created a cell-laden 3D tissue analog to study indirect crosstalk between these two cell types and their direct synergistic effect when cultured on a 3D scaffold. The cell-specific role of osteoclast differentiation was investigated through osteoblast- and proinflammatory macrophage-specific feedback studies. The results suggested that when macrophages were exposed to osteoblasts-derived conditioned media from the mineralized matrix, the M1 macrophages tended to maintain their proinflammatory phenotype. Further, when osteoblasts were exposed to secretions from proinflammatory macrophages, they demonstrated elevated receptor activator of nuclear factor-κB ligand (RANKL) expression and decreased alkaline phosphate (ALP) activities compared to osteoblasts exposed to only osteogenic media. In addition, the upregulation of tumor necrosis factor-alpha (TNF-α) and c-Fos in proinflammatory macrophages within the 3D matrix indirectly increased the RANKL expression and reduced the ALP activity of osteoblasts, promoting osteoclastogenesis. The contact coculturing with osteoblast and proinflammatory macrophages within the 3D matrix demonstrated that the proinflammatory markers (TNF-α and interleukin-1β) expressions were upregulated. In contrast, anti-inflammatory markers (c-c motif chemokine ligand 18 [CCL18]) were downregulated, and osteoclastogenic markers (TNF receptor associated factor 6 [TRAF6] and acid phosphatase 5, tartrate resistant [ACP5]) were unchanged. The data suggested that the osteoblasts curbed the osteoclastogenic differentiation of macrophages while macrophages still preserved their proinflammatory lineages. The osteoblast within the 3D coculture demonstrated increased ALP activity and did not express RANKL significantly different than the osteoblast cultured within a 3D collagen matrix without macrophages. Contact coculturing has an anabolic effect on bone tissue in a bacteria-derived inflammatory environment.
Collapse
Affiliation(s)
- Diego Jacho
- Department of Bioengineering, University of Toledo, Toledo, Ohio, USA
| | | | - Raquel Osorio
- Department of Dentistry, University of Granada, Colegio Máximo de Cartuja, Granada, Spain
- Instituto de Investigación Biosanitaria ibs, University of Granada, Granada, Spain
| | - Manuel Toledano
- Department of Dentistry, University of Granada, Colegio Máximo de Cartuja, Granada, Spain
- Instituto de Investigación Biosanitaria ibs, University of Granada, Granada, Spain
| | - Agustín Rabino
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
19
|
Costa-Verdera H, Unzu C, Valeri E, Adriouch S, González Aseguinolaza G, Mingozzi F, Kajaste-Rudnitski A. Understanding and Tackling Immune Responses to Adeno-Associated Viral Vectors. Hum Gene Ther 2023; 34:836-852. [PMID: 37672519 DOI: 10.1089/hum.2023.119] [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] [Indexed: 09/08/2023] Open
Abstract
As the clinical experience in adeno-associated viral (AAV) vector-based gene therapies is expanding, the necessity to better understand and control the host immune responses is also increasing. Immunogenicity of AAV vectors in humans has been linked to several limitations of the platform, including lack of efficacy due to antibody-mediated neutralization, tissue inflammation, loss of transgene expression, and in some cases, complement activation and acute toxicities. Nevertheless, significant knowledge gaps remain in our understanding of the mechanisms of immune responses to AAV gene therapies, further hampered by the failure of preclinical animal models to recapitulate clinical findings. In this review, we focus on the current knowledge regarding immune responses, spanning from innate immunity to humoral and adaptive responses, triggered by AAV vectors and how they can be mitigated for safer, durable, and more effective gene therapies.
Collapse
Affiliation(s)
- Helena Costa-Verdera
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, Milan, Italy
| | - Carmen Unzu
- DNA and RNA Medicine Division, CIMA, Universidad de Navarra, IdisNA, Pamplona, Spain
| | - Erika Valeri
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, Milan, Italy
| | - Sahil Adriouch
- University of Rouen, INSERM, U1234, Pathophysiology Autoimmunity and Immunotherapy (PANTHER), Normandie University, Rouen, France
| | - Gloria González Aseguinolaza
- DNA and RNA Medicine Division, CIMA, Universidad de Navarra, IdisNA, Pamplona, Spain
- Vivet Therapeutics S.L., Pamplona, Spain; and
| | | | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCSS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
20
|
Yang M, Peng B, Zhuang Q, Li J, Zhang P, Liu H, Zhu Y, Ming Y. Machine learning-based investigation of the relationship between immune status and left ventricular hypertrophy in patients with end-stage kidney disease. Front Cardiovasc Med 2023; 10:1187965. [PMID: 37273870 PMCID: PMC10233114 DOI: 10.3389/fcvm.2023.1187965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023] Open
Abstract
Background Left ventricular hypertrophy (LVH) is the most frequent cardiac complication among end-stage kidney disease (ESKD) patients, which has been identified as predictive of adverse outcomes. Emerging evidence has suggested that immune system is implicated in the development of cardiac hypertrophy in multiple diseases. We applied machine learning models to exploring the relation between immune status and LVH in ESKD patients. Methods A cohort of 506 eligible patients undergoing immune status assessment and standard echocardiography simultaneously in our center were retrospectively analyzed. The association between immune parameters and the occurrence of LVH were evaluated through univariate and multivariate logistic analysis. To develop a predictive model, we utilized four distinct modeling approaches: support vector machine (SVM), logistic regression (LR), multi-layer perceptron (MLP), and random forest (RF). Results In comparison to the non-LVH group, ESKD patients with LVH exhibited significantly impaired immune function, as indicated by lower cell counts of CD3+ T cells, CD4+ T cells, CD8+ T cells, and B cells. Additionally, multivariable Cox regression analysis revealed that a decrease in CD3+ T cell count was an independent risk factor for LVH, while a decrease in NK cell count was associated with the severity of LVH. The RF model demonstrated superior performance, with an average area under the curve (AUC) of 0.942. Conclusion Our findings indicate a strong association between immune parameters and LVH in ESKD patients. Moreover, the RF model exhibits excellent predictive ability in identifying ESKD patients at risk of developing LVH. Based on these results, immunomodulation may represent a promising approach for preventing and treating this disease.
Collapse
Affiliation(s)
- Min Yang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Bo Peng
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Quan Zhuang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Junhui Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Pengpeng Zhang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Hong Liu
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yi Zhu
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| |
Collapse
|
21
|
Ciaunica A, Shmeleva EV, Levin M. The brain is not mental! coupling neuronal and immune cellular processing in human organisms. Front Integr Neurosci 2023; 17:1057622. [PMID: 37265513 PMCID: PMC10230067 DOI: 10.3389/fnint.2023.1057622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/18/2023] [Indexed: 06/03/2023] Open
Abstract
Significant efforts have been made in the past decades to understand how mental and cognitive processes are underpinned by neural mechanisms in the brain. This paper argues that a promising way forward in understanding the nature of human cognition is to zoom out from the prevailing picture focusing on its neural basis. It considers instead how neurons work in tandem with other type of cells (e.g., immune) to subserve biological self-organization and adaptive behavior of the human organism as a whole. We focus specifically on the immune cellular processing as key actor in complementing neuronal processing in achieving successful self-organization and adaptation of the human body in an ever-changing environment. We overview theoretical work and empirical evidence on "basal cognition" challenging the idea that only the neuronal cells in the brain have the exclusive ability to "learn" or "cognize." The focus on cellular rather than neural, brain processing underscores the idea that flexible responses to fluctuations in the environment require a carefully crafted orchestration of multiple cellular and bodily systems at multiple organizational levels of the biological organism. Hence cognition can be seen as a multiscale web of dynamic information processing distributed across a vast array of complex cellular (e.g., neuronal, immune, and others) and network systems, operating across the entire body, and not just in the brain. Ultimately, this paper builds up toward the radical claim that cognition should not be confined to one system alone, namely, the neural system in the brain, no matter how sophisticated the latter notoriously is.
Collapse
Affiliation(s)
- Anna Ciaunica
- Centre for Philosophy of Science, Faculty of Science, University of Lisbon, Lisbon, Portugal
- Faculty of Brain Sciences, Institute of Cognitive Neuroscience, University College London, London, United Kingdom
| | - Evgeniya V. Shmeleva
- Department of Biology, Tufts University, Medford, MA, United States
- Allen Discovery Center, Tufts University, Medford, MA, United States
| | - Michael Levin
- Department of Biology, Tufts University, Medford, MA, United States
- Allen Discovery Center, Tufts University, Medford, MA, United States
| |
Collapse
|
22
|
Barnable P, Mukhopadhyay S, Kizima L, Kumar N, Plagianos M, Mehandru S, Teleshova N. Ex Vivo Colonic Tissue Susceptibility to HIV-1 in Cisgender Men and Women. AIDS Res Hum Retroviruses 2023; 40:28-36. [PMID: 37002886 PMCID: PMC10790552 DOI: 10.1089/aid.2022.0133] [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] [Indexed: 04/04/2023] Open
Abstract
The biology of HIV-1 acquisition through unprotected receptive anal intercourse is understudied. Considering that sex hormones are implicated in intestinal physiology, pathology, and HIV acquisition and pathogenesis, we explored links between sex hormones, ex vivo HIV-1BaL infection of colonic mucosa, and candidate biomarkers of susceptibility to HIV-1 (CD4+ T cell frequencies and immune mediators) in cisgender women and men. No consistent significant associations between sex hormone concentrations and ex vivo tissue infection with HIV-1BaL were detected. In men, serum estradiol (E2) concentrations were positively associated with tissue proinflammatory mediators (IL17A, GM-CSF, IFNγ, TNFα, and MIG/CXCL9) and serum testosterone concentrations were negatively associated with frequencies of activated CD4+ T cells (CD4+CCR5+, CD4+HLA-DR+, and CD4+CD38+HLA-DR+). In women, the only significant interactions were positive associations between progesterone (P4)/E2 ratios and tissue ILRA concentrations and between P4/E2 ratios and frequencies of tissue CD4+α4β7high+ T cells. The study did not reveal relationships between biological sex or phase of the menstrual cycle and ex vivo tissue HIV-1BaL infection and tissue immune mediators. A comparison of CD4+ T cell frequencies between study groups revealed a higher frequency of tissue CD4+α4β7high+ T cells in women versus men. In contrast, higher frequencies of tissue CD4+CD103+ T cells were detected in men versus women in the follicular phase of the menstrual cycle. Overall, the study identified associations between systemic sex hormone concentrations, biological sex, and tissue candidate biomarkers of susceptibility to HIV-1. The significance of these results for tissue susceptibility to HIV-1 and early HIV-1 pathogenesis warrants further investigation.
Collapse
Affiliation(s)
- Patrick Barnable
- Center for Biomedical Research, Population Council, New York, New York, USA
| | | | - Larisa Kizima
- Center for Biomedical Research, Population Council, New York, New York, USA
| | - Narender Kumar
- Center for Biomedical Research, Population Council, New York, New York, USA
| | - Marlena Plagianos
- Center for Biomedical Research, Population Council, New York, New York, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Dr. Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Natalia Teleshova
- Center for Biomedical Research, Population Council, New York, New York, USA
| |
Collapse
|
23
|
von Gunten S, Schneider C, Imamovic L, Gorochov G. Antibody diversity in IVIG: Therapeutic opportunities for novel immunotherapeutic drugs. Front Immunol 2023; 14:1166821. [PMID: 37063852 PMCID: PMC10090664 DOI: 10.3389/fimmu.2023.1166821] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Significant progress has been made in the elucidation of human antibody repertoires. Furthermore, non-canonical functions of antibodies have been identified that reach beyond classical functions linked to protection from pathogens. Polyclonal immunoglobulin preparations such as IVIG and SCIG represent the IgG repertoire of the donor population and will likely remain the cornerstone of antibody replacement therapy in immunodeficiencies. However, novel evidence suggests that pooled IgA might promote orthobiotic microbial colonization in gut dysbiosis linked to mucosal IgA immunodeficiency. Plasma-derived polyclonal IgG and IgA exhibit immunoregulatory effects by a diversity of different mechanisms, which have inspired the development of novel drugs. Here we highlight recent insights into IgG and IgA repertoires and discuss potential implications for polyclonal immunoglobulin therapy and inspired drugs.
Collapse
Affiliation(s)
- Stephan von Gunten
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- *Correspondence: Stephan von Gunten,
| | | | - Lejla Imamovic
- Sorbonne Université, Inserm, Assistance Publique Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Paris, France
| | - Guy Gorochov
- Sorbonne Université, Inserm, Assistance Publique Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
24
|
Cohen IR, Marron A. Evolution is driven by natural autoencoding: reframing species, interaction codes, cooperation and sexual reproduction. Proc Biol Sci 2023; 290:20222409. [PMID: 36855872 PMCID: PMC9975652 DOI: 10.1098/rspb.2022.2409] [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] [Indexed: 03/02/2023] Open
Abstract
The continuity of life and its evolution, we proposed, emerge from an interactive group process manifested in networks of interaction. We term this process survival of the fitted. Here, we reason that survival of the fitted results from a natural computational process we term natural autoencoding. Natural autoencoding works by retaining repeating biological interactions while non-repeatable interactions disappear. (i) We define a species by its species interaction code, which consists of a compact description of the repeating interactions of species organisms with their external and internal environments. Species interaction codes are descriptions recorded in the biological infrastructure that enables repeating interactions. Encoding and decoding are interwoven. (ii) Evolution proceeds by natural autoencoding of sustained changes in species interaction codes. DNA is only one element in natural autoencoding. (iii) Natural autoencoding accounts for the paradox of genome randomization in sexual reproduction-recombined genomes are analogous to the diversified inputs required for artificial autoencoding. The increase in entropy generated by genome randomization compensates for the decrease in entropy generated by organized life. (iv) Natural autoencoding and artificial autoencoding algorithms manifest defined similarities and differences. Recognition of the importance of fittedness could well serve the future of a humanly livable biosphere.
Collapse
Affiliation(s)
- Irun R. Cohen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Assaf Marron
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
25
|
Swiatczak B. Evolution within the body: the rise and fall of somatic Darwinism in the late nineteenth century. HISTORY AND PHILOSOPHY OF THE LIFE SCIENCES 2023; 45:8. [PMID: 36862350 DOI: 10.1007/s40656-023-00566-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/25/2022] [Indexed: 06/18/2023]
Abstract
Originating in the work of Ernst Haeckel and Wilhelm Preyer, and advanced by a Prussian embryologist, Wilhelm Roux, the idea of struggle for existence between body parts helped to establish a framework, in which population cell dynamics rather than a predefined harmony guides adaptive changes in an organism. Intended to provide a causal-mechanical view of functional adjustments in body parts, this framework was also embraced later by early pioneers of immunology to address the question of vaccine effectiveness and pathogen resistance. As an extension of these early efforts, Elie Metchnikoff established an evolutionary vision of immunity, development, pathology, and senescence, in which phagocyte-driven selection and struggle promote adaptive changes in an organism. Despite its promising start, the idea of somatic evolution lost its appeal at the turn of the twentieth century giving way to a vision, in which an organism operates as a genetically uniform, harmonious entity.
Collapse
Affiliation(s)
- Bartlomiej Swiatczak
- Department of History of Science and Scientific Archaeology, University of Science and Technology of China, 96 Jinzhai Rd. 230026, Hefei, P. R. China.
| |
Collapse
|
26
|
Zhang X, Liu J, Yang X, Jiao W, Shen C, Zhao X, Wang Y. High expression of COL6A1 predicts poor prognosis and response to immunotherapy in bladder cancer. Cell Cycle 2023; 22:610-618. [PMID: 36474424 PMCID: PMC9928451 DOI: 10.1080/15384101.2022.2154551] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular matrix (ECM), as an important framework for tumor microenvironment, plays important roles in many critical processes, including tumor growth, invasion, immune suppression, and drug resistance. However, few biomarkers of ECM-related genes (ERGs) have been developed for prognosis prediction and clinical treatment of bladder cancer (BC) patients. Bioinformatics analysis and LC-MS/MS analysis were used to screen differentially expressed ERGs in BC. Multivariate Cox regression analysis and Lasso regression analysis were used to construct and validate an ERGs-based prognostic prediction model for BC. Immunohistochemistry was used to detect the protein expression of hub gene-COL6A1 in BC patients. Using bioinformatics analysis from The Cancer Genome Atlas (TCGA) database and proteomic analysis from our BC cohort, we constructed and validated an effective prognostic prediction model for BC patients based on four differentially expressed ERGs (MAP1B, FBN1, COL6A1, and MFAP5). Moreover, we identified human collagen VI-COL6A1 was a hub gene in this prognostic prediction model and found that COL6A1 was closely related to malignancy progression, prognosis, and response to PD-1 inhibitor immunotherapy in BC. Our findings highlight the satisfactory predictive value of ECM-related prognostic models in BC and suggested that COL6A1 may be a potential biomarker in predicting malignant progression, prognosis, and efficacy of immunotherapy in BC.
Collapse
Affiliation(s)
- Xuezhou Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jing Liu
- Department of Research Management and International Cooperation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xuecheng Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wei Jiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chengquan Shen
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xinzhao Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China,CONTACT Yonghua Wang Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
27
|
Agarwood Pill Enhances Immune Function in Cyclophosphamide-induced Immunosuppressed Mice. BIOTECHNOL BIOPROC E 2023. [DOI: 10.1007/s12257-022-0345-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
28
|
Daguano Gastaldi V, Bh Wilke J, Weidinger CA, Walter C, Barnkothe N, Teegen B, Luessi F, Stöcker W, Lühder F, Begemann M, Zipp F, Nave KA, Ehrenreich H. Factors predisposing to humoral autoimmunity against brain-antigens in health and disease: Analysis of 49 autoantibodies in over 7000 subjects. Brain Behav Immun 2023; 108:135-147. [PMID: 36323361 DOI: 10.1016/j.bbi.2022.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/24/2022] [Accepted: 10/22/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Circulating autoantibodies (AB) against brain-antigens, often deemed pathological, receive increasing attention. We assessed predispositions and seroprevalence/characteristics of 49 AB in > 7000 individuals. METHODS Exploratory cross-sectional cohort study, investigating deeply phenotyped neuropsychiatric patients and healthy individuals of GRAS Data Collection for presence/characteristics of 49 brain-directed serum-AB. Predispositions were evaluated through GWAS of NMDAR1-AB carriers, analyses of immune check-point genotypes, APOE4 status, neurotrauma. Chi-square, Fisher's exact tests and logistic regression analyses were used. RESULTS Study of N = 7025 subjects (55.8 % male; 41 ± 16 years) revealed N = 1133 (16.13 %) carriers of any AB against 49 defined brain-antigens. Overall, age dependence of seroprevalence (OR = 1.018/year; 95 % CI [1.015-1.022]) emerged, but no disease association, neither general nor with neuropsychiatric subgroups. Males had higher AB seroprevalence (OR = 1.303; 95 % CI [1.144-1.486]). Immunoglobulin class (N for IgM:462; IgA:487; IgG:477) and titers were similar. Abundant were NMDAR1-AB (7.7 %). Low seroprevalence (1.25 %-0.02 %) was seen for most AB (e.g., amphiphysin, KCNA2, ARHGAP26, GFAP, CASPR2, MOG, Homer-3, KCNA1, GLRA1b, GAD65). Non-detectable were others. GWAS of NMDAR1-AB carriers revealed three genome-wide significant SNPs, two intergenic, one in TENM3, previously autoimmune disease-associated. Targeted analysis of immune check-point genotypes (CTLA4, PD1, PD-L1) uncovered effects on humoral anti-brain autoimmunity (OR = 1.55; 95 % CI [1.058-2.271]) and disease likelihood (OR = 1.43; 95 % CI [1.032-1.985]). APOE4 carriers (∼19 %) had lower seropositivity (OR = 0.766; 95 % CI [0.625-0.933]). Neurotrauma predisposed to NMDAR1-AB seroprevalence (IgM: OR = 1.599; 95 % CI [1.022-2.468]). CONCLUSIONS Humoral autoimmunity against brain-antigens, frequent across health and disease, is predicted by age, gender, genetic predisposition, and brain injury. Seroprevalence, immunoglobulin class, or titers do not predict disease.
Collapse
Affiliation(s)
- Vinicius Daguano Gastaldi
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Justus Bh Wilke
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Cosima A Weidinger
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Carolin Walter
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Nadine Barnkothe
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Bianca Teegen
- Institute for Experimental Immunology, Affiliated to Euroimmun, Lübeck, Germany
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine‑Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Winfried Stöcker
- Institute for Experimental Immunology, Affiliated to Euroimmun, Lübeck, Germany
| | - Fred Lühder
- Institute of Neuroimmunology and Multiple Sclerosis Research, University Medical Center, of the Georg August University, Göttingen, Germany
| | - Martin Begemann
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine‑Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany.
| |
Collapse
|
29
|
Grossman Z, Meyerhans A, Bocharov G. An integrative systems biology view of host-pathogen interactions: The regulation of immunity and homeostasis is concomitant, flexible, and smart. Front Immunol 2023; 13:1061290. [PMID: 36761169 PMCID: PMC9904014 DOI: 10.3389/fimmu.2022.1061290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/28/2022] [Indexed: 01/26/2023] Open
Abstract
The systemic bio-organization of humans and other mammals is essentially "preprogrammed", and the basic interacting units, the cells, can be crudely mapped into discrete sets of developmental lineages and maturation states. Over several decades, however, and focusing on the immune system, we and others invoked evidence - now overwhelming - suggesting dynamic acquisition of cellular properties and functions, through tuning, re-networking, chromatin remodeling, and adaptive differentiation. The genetically encoded "algorithms" that govern the integration of signals and the computation of new states are not fully understood but are believed to be "smart", designed to enable the cells and the system to discriminate meaningful perturbations from each other and from "noise". Cellular sensory and response properties are shaped in part by recurring temporal patterns, or features, of the signaling environment. We compared this phenomenon to associative brain learning. We proposed that interactive cell learning is subject to selective pressures geared to performance, allowing the response of immune cells to injury or infection to be progressively coordinated with that of other cell types across tissues and organs. This in turn is comparable to supervised brain learning. Guided by feedback from both the tissue itself and the neural system, resident or recruited antigen-specific and innate immune cells can eradicate a pathogen while simultaneously sustaining functional homeostasis. As informative memories of immune responses are imprinted both systemically and within the targeted tissues, it is desirable to enhance tissue preparedness by incorporating attenuated-pathogen vaccines and informed choice of tissue-centered immunomodulators in vaccination schemes. Fortunately, much of the "training" that a living system requires to survive and function in the face of disturbances from outside or within is already incorporated into its design, so it does not need to deep-learn how to face a new challenge each time from scratch. Instead, the system learns from experience how to efficiently select a built-in strategy, or a combination of those, and can then use tuning to refine its organization and responses. Efforts to identify and therapeutically augment such strategies can take advantage of existing integrative modeling approaches. One recently explored strategy is boosting the flux of uninfected cells into and throughout an infected tissue to rinse and replace the infected cells.
Collapse
Affiliation(s)
- Zvi Grossman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow, Russia
- Institute of Computer Science and Mathematical Modeling, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
30
|
Effenberger M, Widjaja AA, Grabherr F, Schaefer B, Grander C, Mayr L, Schwaerzler J, Enrich B, Moser P, Fink J, Pedrini A, Jaschke N, Kirchmair A, Pfister A, Hausmann B, Bale R, Putzer D, Zoller H, Schafer S, Pjevac P, Trajanoski Z, Oberhuber G, Adolph T, Cook S, Tilg H. Interleukin-11 drives human and mouse alcohol-related liver disease. Gut 2023; 72:168-179. [PMID: 35365572 DOI: 10.1136/gutjnl-2021-326076] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/18/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Alcoholic hepatitis (AH) reflects acute exacerbation of alcoholic liver disease (ALD) and is a growing healthcare burden worldwide. Interleukin-11 (IL-11) is a profibrotic, proinflammatory cytokine with increasingly recognised toxicities in parenchymal and epithelial cells. We explored IL-11 serum levels and their prognostic value in patients suffering from AH and cirrhosis of various aetiology and experimental ALD. DESIGN IL-11 serum concentration and tissue expression was determined in a cohort comprising 50 patients with AH, 110 patients with cirrhosis and 19 healthy volunteers. Findings were replicated in an independent patient cohort (n=186). Primary human hepatocytes exposed to ethanol were studied in vitro. Ethanol-fed wildtype mice were treated with a neutralising murine IL-11 receptor-antibody (anti-IL11RA) and examined for severity signs and markers of ALD. RESULTS IL-11 serum concentration and hepatic expression increased with severity of liver disease, mostly pronounced in AH. In a multivariate Cox-regression, a serum level above 6.4 pg/mL was a model of end-stage liver disease independent risk factor for transplant-free survival in patients with compensated and decompensated cirrhosis. In mice, severity of alcohol-induced liver inflammation correlated with enhanced hepatic IL-11 and IL11RA expression. In vitro and in vivo, anti-IL11RA reduced pathogenic signalling pathways (extracellular signal-regulated kinases, c-Jun N-terminal kinase, NADPH oxidase 4) and protected hepatocytes and murine livers from ethanol-induced inflammation and injury. CONCLUSION Pathogenic IL-11 signalling in hepatocytes plays a crucial role in the pathogenesis of ALD and could serve as an independent prognostic factor for transplant-free survival. Blocking IL-11 signalling might be a therapeutic option in human ALD, particularly AH.
Collapse
Affiliation(s)
- Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Anissa A Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Benedikt Schaefer
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Julian Schwaerzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Moser
- INNPATH, Innsbruck Medical University Hospital, Innsbruck, Austria
| | - Julia Fink
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Alisa Pedrini
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Nikolai Jaschke
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander Kirchmair
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexandra Pfister
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Reto Bale
- Department of Radiology, Section of Interventional Oncology-Microinvasive Therapy (SIP), Medical University of Innsbruck, Innsbruck, Austria
| | - Daniel Putzer
- Department of Radiology, Section of Interventional Oncology-Microinvasive Therapy (SIP), Medical University of Innsbruck, Innsbruck, Austria
| | - Heinz Zoller
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schafer
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Petra Pjevac
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Medical University of Vienna, University of Vienna, Vienna, Austria
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Oberhuber
- INNPATH, Innsbruck Medical University Hospital, Innsbruck, Austria
| | - Timon Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Stuart Cook
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London, UK
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Smith LE, Chagwena DT, Bourke C, Robertson R, Fernando S, Tavengwa NV, Cairns J, Ndhlela T, Matumbu E, Brown T, Datta K, Mutasa B, Tengende A, Chidhanguro D, Langhaug L, Makanza M, Chasekwa B, Mutasa K, Swann J, Kelly P, Ntozini R, Prendergast A. Child Health, Agriculture and Integrated Nutrition (CHAIN): protocol for a randomised controlled trial of improved infant and young child feeding in rural Zimbabwe. BMJ Open 2022; 12:e056435. [PMID: 36585147 PMCID: PMC9809274 DOI: 10.1136/bmjopen-2021-056435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/28/2022] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Over one-quarter of children in sub-Saharan Africa are stunted; however, commercial supplements only partially meet child nutrient requirements, cannot be sustainably produced, and do not resolve physiological barriers to adequate nutrition (eg, inflammation, microbiome dysbiosis and metabolic dysfunction). Redesigning current infant and young child feeding (IYCF) interventions using locally available foods to improve intake, uptake and utilisation of nutrients could ameliorate underlying pathogenic pathways and improve infant growth during the critical period of complementary feeding, to reduce the global burden of stunting. METHODS AND ANALYSIS Child Health Agriculture Integrated Nutrition is an open-label, individual household randomised trial comparing the effects of IYCF versus 'IYCF-plus' on nutrient intake during infancy. The IYCF intervention comprises behaviour change modules to promote infant nutrition delivered by community health workers, plus small-quantity lipid-based nutrient supplements from 6 to 12 months of age which previously reduced stunting at 18 months of age by ~20% in rural Zimbabwe. The 'IYCF-plus' intervention provides these components plus powdered NUA-45 biofortified sugar beans, whole egg powder, moringa leaf powder and provitamin A maize. The trial will enrol 192 infants between 5 and 6 months of age in Shurugwi district, Zimbabwe. Research nurses will collect data plus blood, urine and stool samples at baseline (5-6 months of age) and endline (9-11 months of age). The primary outcome is energy intake, measured by multipass 24-hour dietary recall at 9-11 months of age. Secondary outcomes include nutrient intake, anthropometry and haemoglobin concentration. Nested laboratory substudies will evaluate the gut microbiome, environmental enteric dysfunction, metabolic phenotypes and innate immune function. Qualitative substudies will explore the acceptability and feasibility of the IYCF-plus intervention among participants and community stakeholders, and the effects of migration on food production and consumption. ETHICS AND DISSEMINATION This trial is registered at ClinicalTrials.gov (NCT04874688) and was approved by the Medical Research Council of Zimbabwe (MRCZ/A/2679) with the final version 1.4 approved on 20 August 2021, following additional amendments. Dissemination of trial results will be conducted through the Community Engagement Advisory Board in the study district and through national-level platforms. TRIAL REGISTRATION NUMBER NCT04874688.
Collapse
Affiliation(s)
- Laura E Smith
- Public and Ecosystem Health, Cornell University, Ithaca, New York, USA
| | - Dexter T Chagwena
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Nutrition, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Claire Bourke
- Blizard Institute, Queen Mary University, London, UK
| | | | - Shamiso Fernando
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V Tavengwa
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | | | - Exhibit Matumbu
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | | | - Batsirai Mutasa
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Alice Tengende
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Dzivaidzo Chidhanguro
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Lisa Langhaug
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Maggie Makanza
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Bernard Chasekwa
- Nutrition, Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jonathan Swann
- University of Southampton Faculty of Medicine, Southampton, UK
| | - Paul Kelly
- Barts and The London School of Medicine, London, UK
| | - Robert Ntozini
- Biostatistics & IT, Zvitambo Institute, Harare, Zimbabwe
| | | |
Collapse
|
32
|
Thin KA, Angsuwatcharakon P, Edwards SW, Mutirangura A, Puttipanyalears C. Upregulation of p16INK4A in Peripheral White Blood Cells as a Novel Screening Marker for Colorectal Carcinoma. Asian Pac J Cancer Prev 2022; 23:3753-3761. [PMID: 36444588 PMCID: PMC9930939 DOI: 10.31557/apjcp.2022.23.11.3753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE Screening of colorectal cancer (CRC) is important for the early detection. CRC is relating to aging and immuno-senescence. One such senescent marker is p16INK4A expression in immune cells. The objective of the study is to investigate the protein expression of p16INK4A in peripheral white blood cells as a screening marker for colorectal cancer. METHODS A case-control studies were conducted. Cases were patients with colorectal cancer and controls were matched with cases based on age and sex. Peripheral blood was collected from patients and controls and the protein p16INK4A was measured with immunofluorescent techniques. The p16INK4A levels from cases and controls were evaluated using ROC analysis to be used as a screening marker in CRC patients. Mean fluorescent intensity of p16INK4A of cases and controls were analyzed in CD45+, CD3+ or CD14+ cells. The p16INK4A levels of cases were also correlated with clinical data. RESULT Statistically significant increased expression of p16INK4A levels were found in cases compared to controls. p16INK4A in peripheral immune cells had 78% sensitivity and 71% specificity which can possibly be used as a diagnosis tool for colorectal cancer. P16INK4A-positive cell percentage and mean florescent intensity were significantly higher in CD45+ cells, CD3 positive cells and CD14 positive cells. No significant correlation was observed with the clinical data and p16INK4A level of CRC patients. CONCLUSION The significant increase of p16 INK4A expression level in peripheral immune cells represents potential for use as a CRC screening marker.
Collapse
Affiliation(s)
- Khin Aye Thin
- Joint PhD Program in Biomedical Sciences and Biotechnology between Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand and Institute of Integrative Biology, University of Liverpool, Liverpool, L7 8TX, United Kingdom.
| | | | - Steven W Edwards
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L7 8TX, United Kingdom.
| | - Apiwat Mutirangura
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand. ,Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Charoenchai Puttipanyalears
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand. ,Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand. ,For Correspondence:
| |
Collapse
|
33
|
Mileto SJ, Hutton ML, Walton SL, Das A, Ioannidis LJ, Ketagoda D, Quinn KM, Denton KM, Hansen DS, Lyras D. Bezlotoxumab prevents extraintestinal organ damage induced by Clostridioides difficile infection. Gut Microbes 2022; 14:2117504. [PMID: 36045589 PMCID: PMC9450906 DOI: 10.1080/19490976.2022.2117504] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Clostridioides difficile is the most common cause of infectious antibiotic-associated diarrhea, with disease mediated by two major toxins TcdA and TcdB. In severe cases, systemic disease complications may arise, resulting in fatal disease. Systemic disease in animal models has been described, with thymic damage an observable consequence of severe disease in mice. Using a mouse model of C. difficile infection, we examined this disease phenotype, focussing on the thymus and serum markers of systemic disease. The efficacy of bezlotoxumab, a monoclonal TcdB therapeutic, to prevent toxin mediated systemic disease complications was also examined. C. difficile infection causes toxin-dependent thymic damage and CD4+CD8+ thymocyte depletion in mice. These systemic complications coincide with changes in biochemical markers of liver and kidney function, including increased serum urea and creatinine, and hypoglycemia. Administration of bezlotoxumab during C. difficile infection prevents systemic disease and thymic atrophy, without blocking gut damage, suggesting the leakage of gut contents into circulation may influence systemic disease. As the thymus has such a crucial role in T cell production and immune system development, these findings may have important implications in relapse of C. difficile disease and impaired immunity during C. difficile infection. The prevention of thymic atrophy and reduced systemic response following bezlotoxumab treatment, without altering colonic damage, highlights the importance of systemic disease in C. difficile infection, and provides new insights into the mechanism of action for this therapeutic.Abbreviations: Acute kidney injury (AKI); Alanine Transaminase (ALT); Aspartate Aminotransferase (AST); C. difficile infection (CDI); chronic kidney disease (CKD); combined repetitive oligo-peptides (CROPS); cardiovascular disease (CVD); Double positive (DP); hematoxylin and eosin (H&E); immunohistochemical (IHC); multiple organ dysfunction syndrome (MODS); phosphate buffered saline (PBS); standard error of the mean (SEM); surface layer proteins (SLP); Single positive (SP); wild-type (WT).
Collapse
Affiliation(s)
- Steven J. Mileto
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Melanie L. Hutton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Sarah L. Walton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Antariksh Das
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Lisa J. Ioannidis
- Walter and Eliza Hall Insitiute, Infectious Diseases and Immune Defence Division, Parkville, Australia,Department of Medical Biology, the University of Melbourne, Parkville, Australia
| | - Don Ketagoda
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Kylie M. Quinn
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia,RMIT University School of Biomedical and Health Sciences, Chronic Inflammatory and Infectious Diseases Program, Bundoora, Australia
| | - Kate M. Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Diana S. Hansen
- Walter and Eliza Hall Insitiute, Infectious Diseases and Immune Defence Division, Parkville, Australia,Department of Medical Biology, the University of Melbourne, Parkville, Australia
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia,CONTACT Dena Lyras Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria3800, Australia
| |
Collapse
|
34
|
Beasley GM, Brown MC, Farrow NE, Landa K, Al-Rohil RN, Selim MA, Therien AD, Jung SH, Gao J, Boczkowski D, Holl EK, Salama AKS, Bigner DD, Gromeier M, Nair SK. Multimodality analysis confers a prognostic benefit of a T-cell infiltrated tumor microenvironment and peripheral immune status in patients with melanoma. J Immunother Cancer 2022; 10:e005052. [PMID: 36175036 PMCID: PMC9528663 DOI: 10.1136/jitc-2022-005052] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We previously reported results from a phase 1 study testing intratumoral recombinant poliovirus, lerapolturev, in 12 melanoma patients. All 12 patients received anti-PD-1 systemic therapy before lerapolturev, and 11 of these 12 patients also received anti-PD-1 after lerapolturev. In preclinical models lerapolturev induces intratumoral innate inflammation that engages antitumor T cells. In the current study, prelerapolturev and postlerapolturev tumor biopsies and blood were evaluated for biomarkers of response. METHODS The following analyses were performed on tumor tissue (n=11): (1) flow cytometric assessment of immune cell density, (2) NanoString Digital Spatial profiling of protein and the transcriptome, and (3) bulk RNA sequencing. Immune cell phenotypes and responsiveness to in vitro stimulation, including in vitro lerapolturev challenge, were measured in peripheral blood (n=12). RESULTS Three patients who received anti-PD-1 therapy within 30 days of lerapolturev have a current median progression-free survival (PFS) of 2.3 years and had higher CD8+T cell infiltrates in prelerapolturev tumor biopsies relative to that of 7 patients with median PFS of 1.6 months and lower CD8+T cell infiltrates in prelerapolturev tumor biopsies. In peripheral blood, four patients with PFS 2.3 years (including three that received anti-PD-1 therapy within 30 days before lerapolturev and had higher pretreatment tumor CD8+T cell infiltrates) had significantly higher effector memory (CD8+, CCR7-, CD45RA-) but lower CD8+PD-1+ and CD4+PD-1+ cells compared with eight patients with median PFS 1.6 months. In addition, pretreatment blood from the four patients with median PFS 2.3 years had more potent antiviral responses to in vitro lerapolturev challenge compared with eight patients with median PFS 1.6 months. CONCLUSION An inflamed pretreatment tumor microenvironment, possibly induced by prior anti-PD-1 therapy and a proficient peripheral blood pretreatment innate immune response (antiviral/interferon signaling) to lerapolturev was associated with long term PFS after intratumoral lerapolturev in a small cohort of patients. These findings imply a link between intratumoral T cell inflammation and peripheral immune function. TRIAL REGISTRATION NUMBER NCT03712358.
Collapse
Affiliation(s)
- Georgia M Beasley
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Michael C Brown
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Norma E Farrow
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Karenia Landa
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Rami N Al-Rohil
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | | | - Aaron D Therien
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Sin-Ho Jung
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Junheng Gao
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - David Boczkowski
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Eda K Holl
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - April K S Salama
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Darell D Bigner
- Department of Medicine, Duke University, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Matthias Gromeier
- Department of Medicine, Duke University, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Smita K Nair
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
35
|
van Sleen Y, van der Geest KSM, Reitsema RD, Esen I, Terpstra JH, Raveling-Eelsing E, van der Heiden M, Lieber T, Buisman AM, van Baarle D, Sandovici M, Brouwer E. Humoral and cellular SARS-CoV-2 vaccine responses in patients with giant cell arteritis and polymyalgia rheumatica. RMD Open 2022. [PMCID: PMC9453427 DOI: 10.1136/rmdopen-2022-002479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Objectives Giant cell arteritis (GCA) and polymyalgia rheumatica (PMR) are overlapping autoinflammatory diseases affecting people over 50 years. The diseases are treated with immunosuppressive drugs such as prednisolone, methotrexate, leflunomide and tocilizumab. In this study, we assessed the immunogenicity and safety of SARS-CoV-2 vaccinations in these diseases (based on humoral and cellular immunity). Methods Patients (n=45 GCA, n=33 PMR) visited the outpatient clinic twice: pre-vaccination and 4 weeks after the second dose (BNT162b2 or ChAdOx1 vaccine). Patients with previous SARS-CoV-2 infection were excluded. In both pre-vaccination and post-vaccination samples, anti-Spike antibody concentrations were assessed and compared with age-, sex- and vaccine-matched control groups (n=98). In addition, the frequency of SARS-CoV-2 Spike-specific T-cells was assessed by IFN-γ ELIspot assay, and side effects and disease activity were recorded. Results GCA/PMR patients did not have reduced antibody concentrations compared with controls. However, linear regression analysis revealed a significant association of methotrexate and >10 mg/day prednisolone use with lower antibody concentrations in GCA/PMR patients. Evidence of cellular immunity, as assessed by ELIspot assay, was found in 67% of GCA/PMR patients. Patients using >10 mg/day prednisolone had reduced cellular immunity. Importantly, vaccination did not lead to significant side effects or changes in disease activity. Conclusions SARS-CoV-2 vaccination was safe for GCA/PMR patients and immunogenicity was comparable to other older individuals. However, patients using methotrexate and particularly >10 mg/day prednisolone did show lower vaccine responses, which corroborates findings in other autoinflammatory patient populations. These patients may therefore be at higher risk of (potentially even severe) breakthrough SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Kornelis S M van der Geest
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Rosanne D Reitsema
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Idil Esen
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Janneke H Terpstra
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth Raveling-Eelsing
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Thomas Lieber
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| | - Annemarie M Buisman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
36
|
Floudas A, Smith CM, Tynan O, Neto N, Krishna V, Wade SM, Hanlon M, Cunningham C, Marzaioli V, Canavan M, Fletcher JM, Mullan RH, Cole S, Hao LY, Monaghan MG, Nagpal S, Veale DJ, Fearon U. Distinct stromal and immune cell interactions shape the pathogenesis of rheumatoid and psoriatic arthritis. Ann Rheum Dis 2022; 81:1224-1242. [PMID: 35701153 DOI: 10.1136/annrheumdis-2021-221761] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 05/12/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Immune and stromal cell communication is central in the pathogenesis of rheumatoid arthritis (RA) and psoriatic arthritis (PsA), however, the nature of these interactions in the synovial pathology of the two pathotypes can differ. Identifying immune-stromal cell crosstalk at the site of inflammation in RA and PsA is challenging. This study creates the first global transcriptomic analysis of the RA and PsA inflamed joint and investigates immune-stromal cell interactions in the pathogenesis of synovial inflammation. METHODS Single cell transcriptomic profiling of 178 000 synovial tissue cells from five patients with PsA and four patients with RA, importantly, without prior sorting of immune and stromal cells. This approach enabled the transcriptomic analysis of the intact synovial tissue and identification of immune and stromal cell interactions. State of the art data integration and annotation techniques identified and characterised 18 stromal and 14 immune cell clusters. RESULTS Global transcriptomic analysis of synovial cell subsets identifies actively proliferating synovial T cells and indicates that due to differential λ and κ immunoglobulin light chain usage, synovial plasma cells are potentially not derived from the local memory B cell pool. Importantly, we report distinct fibroblast and endothelial cell transcriptomes indicating abundant subpopulations in RA and PsA characterised by differential transcription factor usage. Using receptor-ligand interactions and downstream target characterisation, we identify RA-specific synovial T cell-derived transforming growth factor (TGF)-β and macrophage interleukin (IL)-1β synergy in driving the transcriptional profile of FAPα+THY1+ invasive synovial fibroblasts, expanded in RA compared with PsA. In vitro characterisation of patient with RA synovial fibroblasts showed metabolic switch to glycolysis, increased adhesion intercellular adhesion molecules 1 expression and IL-6 secretion in response to combined TGF-β and IL-1β treatment. Disrupting specific immune and stromal cell interactions offers novel opportunities for targeted therapeutic intervention in RA and PsA.
Collapse
Affiliation(s)
- Achilleas Floudas
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Conor M Smith
- Translational Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Orla Tynan
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Nuno Neto
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Dublin, Ireland
| | - Vinod Krishna
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Sarah M Wade
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Megan Hanlon
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Clare Cunningham
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Viviana Marzaioli
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Mary Canavan
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Jean M Fletcher
- Translational Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ronan H Mullan
- Department of Rheumatology, Tallaght University Hospital, Trinity College Dublin, Dublin, Ireland
| | - Suzanne Cole
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Ling-Yang Hao
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Michael G Monaghan
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Dublin, Ireland
| | - Sunil Nagpal
- Immunology, Janssen Research & Development, Spring House, PA, USA
| | - Douglas J Veale
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, Clinical Medicine, Trinity Biomedical Science Institute, Dublin, Ireland
- Eular Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Univeristy College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Qin Y, Cai ML, Jin HZ, Huang W, Zhu C, Bozec A, Huang J, Chen Z. Age-associated B cells contribute to the pathogenesis of rheumatoid arthritis by inducing activation of fibroblast-like synoviocytes via TNF-α-mediated ERK1/2 and JAK-STAT1 pathways. Ann Rheum Dis 2022; 81:1504-1514. [PMID: 35760450 DOI: 10.1136/ard-2022-222605] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/17/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Age-associated B cells (ABCs) are a recently identified B cell subset, whose expansion has been increasingly linked to the pathogenesis of autoimmune disorders. This study aimed to investigate whether ABCs are involved in the pathogenesis and underlying mechanisms of rheumatoid arthritis (RA). METHODS ABCs were assessed in collagen-induced arthritis (CIA) mice and patients with RA using flow cytometry. Transcriptomic features of RA ABCs were explored using RNA-seq. Primary fibroblast-like synoviocytes (FLS) derived from the synovial tissue of patients with RA were cocultured with ABCs or ABCs-conditioned medium (ABCsCM). IL-6, MMP-1, MMP-3 and MMP-13 levels in the coculture supernatant were detected by ELISA. Signalling pathways related to ABCs-induced FLS activation were examined using western blotting. RESULTS Increased ABCs levels in the blood, spleen and inflammatory joints of CIA mice were observed. Notably, ABCs were elevated in the blood, synovial fluid and synovial tissue of patients with RA and positively correlated with disease activity. RNA-seq revealed upregulated chemotaxis-related genes in RA ABCs compared with those in naive and memory B cells. Coculture of FLS with RA ABCs or ABCsCM led to an active phenotype of FLS, with increased production of IL-6, MMP-1, MMP-3 and MMP-13. Mechanistically, ABCsCM-derived TNF-α promoted the upregulation of interferon-stimulated genes in FLS, with elevated phosphorylation of ERK1/2 and STAT1. Furthermore, blockage of ERK1/2 and Janus Kinase (JAK)-STAT1 pathways inhibited the activation of FLS induced by ABCsCM. CONCLUSIONS Our results suggest that ABCs contribute to the pathogenesis of RA by inducing the activation of FLS via TNF-α-mediated ERK1/2 and JAK-STAT1 pathways.
Collapse
Affiliation(s)
- Yi Qin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ming-Long Cai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hui-Zhi Jin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Aline Bozec
- Department of Internal Medicine III, Institute for Clinical Immunology University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jingang Huang
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhu Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
38
|
Macaluso F, Marvisi C, Castrignanò P, Pipitone N, Salvarani C. Comparing treatment options for large vessel vasculitis. Expert Rev Clin Immunol 2022; 18:793-805. [PMID: 35714219 DOI: 10.1080/1744666x.2022.2092098] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Giant cell arteritis (GCA) and Takayasu arteritis (TAK) are the major forms of large vessel vasculitis (LVV).Glucocorticoids represent the cornerstone of LVV treatment, however, relapses and recurrences frequently occur when they are tapered or stopped, determining a prolonged exposure to glucocorticoids and a subsequent increased risk of glucocorticoid-related side effects. Therefore, conventional and biologic immunosuppressive drugs have been proposed to obtain a glucocorticoid-sparing effect. AREAS COVERED We searched PubMed® using the keywords "giant cell arteritis/drug therapy" and "Takayasu Arteritis/drug therapy" OR "Takayasu Arteritis/surgery". This review focuses on the management of LVV, based on the current evidence while highlighting the differences in terms of therapeutic management of TAK and GCA. EXPERT OPINION Conventional disease modifying anti-rheumatic drugs, such as methotrexate or azathioprine, are recommended in association to glucocorticoids for selected GCA and all TAK patients. Two randomized placebo-controlled trials recently demonstrated the efficacy of tocilizumab in reducing relapses and cumulative prednisone dosage in GCA patients with newly diagnosed or relapsing disease. Observational evidence and two small randomized controlled trials support the use of TNF-alpha inhibitors and tocilizumab as glucocorticoid-sparing agents in relapsing TAK, albeit high-quality evidence regarding the management of TAK is still lacking.
Collapse
Affiliation(s)
- Federica Macaluso
- Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Department of Precision Medicine, Section of Rheumatology, Università della Campania L Vanvitelli, Naples, Italy
| | - Chiara Marvisi
- Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Rheumatology Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Paola Castrignanò
- Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Rheumatology Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicolò Pipitone
- Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Salvarani
- Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Rheumatology Unit, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
39
|
In-Silico Design of a Multi‑epitope Construct Against Influenza A Based on Nucleoprotein Gene. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10418-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
40
|
Wang D, Wang J, Liu H, Liu M, Yang Y, Zhong S. The Main Structural Unit Elucidation and Immunomodulatory Activity In Vitro of a Selenium-Enriched Polysaccharide Produced by Pleurotus ostreatus. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082591. [PMID: 35458788 PMCID: PMC9027278 DOI: 10.3390/molecules27082591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022]
Abstract
In recent years, the structure of selenium-enriched polysaccharides and their application in immunomodulation have attracted much attention. In previous studies, we extracted and purified a novel selenium-enriched Pleurotus ostreatus polysaccharide called Se-POP-21, but its structure and immunomodulatory activity were still unclear. In this study, the main structural unit formula of Se-POP-21 was characterized by methylation analysis and an NMR experiment. The results showed that the backbone of Se-POP-21 was →[2,6)-α-D-Galp-(1→6)-α-D-Galp-(1]4→2,4)-β-L-Arap-(1→[2,6)-α-D-Galp-(1→6)-α-D-Galp-(1]4→, branched chain of β-D-Manp-(1→ and β-D-Manp-(1→4)-β-L-Arap-(1→ connected with →2,6)-α-D-Galp-(1→ and →2,4)-β-L-Arap-(1→,respectively, through the O-2 bond. In vitro cell experiments indicated that Se-POP-21 could significantly enhance the proliferation and phagocytosis of RAW264.7 cells, upregulate the expression of costimulatory molecules CD80/CD86, and promote RAW264.7 cells to secrete NO, ROS, TNF-α, IL-1β, and IL-6 by activating the NF-κB protein. The results of this study indicate that Se-POP-21 can effectively activate RAW264.7 cells. Thus, it has the potential to be used in immunomodulatory drugs or functional foods.
Collapse
|
41
|
Liang C, Huang T, Zhang X, Rao H, Jin Z, Pan X, Li J, Mo Y, Cai Y, Wu J. cRGD Urokinase Liposomes for Thrombolysis in Rat Model of Acute Pulmonary Microthromboembolism. Drug Des Devel Ther 2022; 16:801-816. [PMID: 35370400 PMCID: PMC8964449 DOI: 10.2147/dddt.s351021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Purpose To study the thrombolytic effect and safety of cRGD urokinase liposomes (cRGD-UK-LIP) in rats with acute pulmonary microthromboembolism (APMTE), and explore the application value of echocardiography (ECHO) in animal models. Patients and Methods Ninety-six SD rats were randomized into 6 groups (16/group): normal control, sham operation, APMTE, normal saline (NS), free urokinase (UK), cRGD-UK-LIP. Four groups (APMTE, NS, UK, cRGD-UK-LIP) of rats were injected with autologous thrombus to induce APMTE. Samples were injected into 3 groups (NS, UK, cRGD-UK-LIP) of rats after modeling. Echocardiography was used to assess right ventricle (RV) function and morphology in rats. Six rats in each group were randomly selected and pulmonary artery pressure (PAP) of them was measured through ECHO-guided transthoracic puncture. Finally, the rats were killed and their tissues were taken for pathological examination. Results Compared with normal control or sham operation group, rats in APMTE group had enlarged RV, decreased RV function, increased PAP, and lung tissue of them showed postthromboembolic appearance. There was no significant difference between NS group and APMTE group. RV morphology and function of rats in the UK group and cRGD-UK-LIP group were better and vessels with residual thrombus in these 2 groups were less than APMTE group, especially in the cRGD-UK-LIP group. In terms of PAP, only cRGD-UK-LIP group was significantly lower than APMTE group. No hyperemia, bleeding and swelling were observed in heart, liver and kidney of rats in each group. Conclusion A rat model of APMTE was successfully established. cRGD-UK-LIP has better thrombolytic effect than free urokinase and it is safe. Echocardiography is not merely an important way to evaluate the morphology and function of RV, transthoracic puncture measurement under the guidance of it can be an effective way to monitor PAP in animal models.
Collapse
Affiliation(s)
- Chunting Liang
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Tongtong Huang
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Xiaofeng Zhang
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Huaqing Rao
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Zhiru Jin
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Xiaoxiong Pan
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Jingtao Li
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Yingying Mo
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Yongzhi Cai
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| | - Ji Wu
- Department of Ultrasonic Medicine, The First Hospital Affiliated to Guangxi Medical University, Nanning, People’s Republic of China
| |
Collapse
|
42
|
Osorio C, Sfera A, Anton JJ, Thomas KG, Andronescu CV, Li E, Yahia RW, Avalos AG, Kozlakidis Z. Virus-Induced Membrane Fusion in Neurodegenerative Disorders. Front Cell Infect Microbiol 2022; 12:845580. [PMID: 35531328 PMCID: PMC9070112 DOI: 10.3389/fcimb.2022.845580] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
A growing body of epidemiological and research data has associated neurotropic viruses with accelerated brain aging and increased risk of neurodegenerative disorders. Many viruses replicate optimally in senescent cells, as they offer a hospitable microenvironment with persistently elevated cytosolic calcium, abundant intracellular iron, and low interferon type I. As cell-cell fusion is a major driver of cellular senescence, many viruses have developed the ability to promote this phenotype by forming syncytia. Cell-cell fusion is associated with immunosuppression mediated by phosphatidylserine externalization that enable viruses to evade host defenses. In hosts, virus-induced immune dysfunction and premature cellular senescence may predispose to neurodegenerative disorders. This concept is supported by novel studies that found postinfectious cognitive dysfunction in several viral illnesses, including human immunodeficiency virus-1, herpes simplex virus-1, and SARS-CoV-2. Virus-induced pathological syncytia may provide a unified framework for conceptualizing neuronal cell cycle reentry, aneuploidy, somatic mosaicism, viral spreading of pathological Tau and elimination of viable synapses and neurons by neurotoxic astrocytes and microglia. In this narrative review, we take a closer look at cell-cell fusion and vesicular merger in the pathogenesis of neurodegenerative disorders. We present a "decentralized" information processing model that conceptualizes neurodegeneration as a systemic illness, triggered by cytoskeletal pathology. We also discuss strategies for reversing cell-cell fusion, including, TMEM16F inhibitors, calcium channel blockers, senolytics, and tubulin stabilizing agents. Finally, going beyond neurodegeneration, we examine the potential benefit of harnessing fusion as a therapeutic strategy in regenerative medicine.
Collapse
Affiliation(s)
- Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Adonis Sfera
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Jonathan J. Anton
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Karina G. Thomas
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Christina V. Andronescu
- Medical Anthropology – Department of Anthropology, Stanford University, Stanford, CA, United States
| | - Erica Li
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Rayan W. Yahia
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Andrea García Avalos
- Universidad Nacional Autónoma de México (UNAM), Facultad de Medicina Campus, Ciudad de Mexico, Mexico
| | - Zisis Kozlakidis
- International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
43
|
Turiello R, Capone M, Morretta E, Monti MC, Madonna G, Azzaro R, Del Gaudio P, Simeone E, Sorrentino A, Ascierto PA, Morello S. Exosomal CD73 from serum of patients with melanoma suppresses lymphocyte functions and is associated with therapy resistance to anti-PD-1 agents. J Immunother Cancer 2022; 10:jitc-2021-004043. [PMID: 35273100 PMCID: PMC8915288 DOI: 10.1136/jitc-2021-004043] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND CD73 is an ectonucleotidase producing the immunosuppressor mediator adenosine. Elevated levels of circulating CD73 in patients with cancer have been associated with disease progression and poor response to immunotherapy. Immunosuppressive pathways associated with exosomes can affect T-cell function and the therapeutic efficacy of anti-programmed cell-death protein 1 (anti-PD-1) therapy. Here, we conducted a retrospective pilot study to evaluate levels of exosomal CD73 before and early during treatment with anti-PD-1 agents in patients with melanoma and its potential contribution to affect T-cell functions and to influence the clinical outcomes of anti-PD-1 monotherapy. METHODS Exosomes were isolated by mini size exclusion chromatography from serum of patients with melanoma (n=41) receiving nivolumab or pembrolizumab monotherapy. Expression of CD73 and programmed death-ligand 1 (PD-L1) were evaluated on exosomes enriched for CD63 by on-bead flow cytometry. The CD73 AMPase activity was evaluated by mass spectrometry, also in the presence of selective inhibitors of CD73. Interferon (IFN)-γ production and granzyme B expression were measured in CD3/28 activated T cells incubated with exosomes in presence of the CD73 substrate AMP. Levels of CD73 and PD-L1 on exosomes were correlated with therapy response. Exosomes isolated from healthy subjects were used as control. RESULTS Isolated exosomes carried CD73 on their surface, which is enzymatically active in producing adenosine. Incubation of exosomes with CD3/28 activated T cells in the presence of AMP resulted in a significant reduction of IFN-γ release, which was reversed by the CD73 inhibitor APCP or by the selective A2A adenosine receptor antagonist ZM241385. Expression levels of exosomal CD73 from serum of patients with melanoma were not significantly different from those in healthy subjects. Early on-treatment, expression levels of both CD73 and PD-L1 on exosomes isolated from patients receiving pembrolizumab or nivolumab monotherapy were significantly increased compared with baseline. Early during therapy exosomal PD-L1 increased in responders, while exosomal CD73 resulted significantly increased in non-responders. CONCLUSIONS CD73 expressed on exosomes from serum of patients with melanoma produces adenosine and contributes to suppress T-cell functions. Early on-treatment, elevated expression levels of exosomal CD73 might affect the response to anti-PD-1 agents in patients with melanoma who failed to respond to therapy.
Collapse
Affiliation(s)
- Roberta Turiello
- Department of Pharmacy, University of Salerno, Fisciano, Italy.,PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, Italy
| | | | - Elva Morretta
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | | | - Gabriele Madonna
- Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | - Rosa Azzaro
- Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | | | - Ester Simeone
- Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| |
Collapse
|
44
|
Grieshaber-Bouyer R, Exner T, Hackert NS, Radtke FA, Jelinsky SA, Halyabar O, Wactor A, Karimizadeh E, Brennan J, Schettini J, Jonsson H, Rao DA, Henderson LA, Müller-Tidow C, Lorenz HM, Wabnitz G, Lederer JA, Hadjipanayis A, Nigrovic PA. Ageing and interferon gamma response drive the phenotype of neutrophils in the inflamed joint. Ann Rheum Dis 2022; 81:805-814. [PMID: 35168946 DOI: 10.1136/annrheumdis-2021-221866] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/02/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Neutrophils are typically the most abundant leucocyte in arthritic synovial fluid. We sought to understand changes that occur in neutrophils as they migrate from blood to joint. METHODS We performed RNA sequencing of neutrophils from healthy human blood, arthritic blood and arthritic synovial fluid, comparing transcriptional signatures with those from murine K/BxN serum transfer arthritis. We employed mass cytometry to quantify protein expression and sought to reproduce the synovial fluid phenotype ex vivo in cultured healthy blood neutrophils. RESULTS Blood neutrophils from healthy donors and patients with active arthritis showed largely similar transcriptional signatures. By contrast, synovial fluid neutrophils exhibited more than 1600 differentially expressed genes. Gene signatures identified a prominent response to interferon gamma (IFN-γ), as well as to tumour necrosis factor, interleukin-6 and hypoxia, in both humans and mice. Mass cytometry confirmed that healthy and arthritic donor blood neutrophils are largely indistinguishable but revealed a range of neutrophil phenotypes in synovial fluid defined by downregulation of CXCR1 and upregulation of FcγRI, HLA-DR, PD-L1, ICAM-1 and CXCR4. Reproduction of key elements of this signature in cultured blood neutrophils required both IFN-γ and prolonged culture. CONCLUSIONS Circulating neutrophils from patients with arthritis resemble those from healthy controls, but joint fluid cells exhibit a network of changes, conserved across species, that implicate IFN-γ response and ageing as complementary drivers of the synovial fluid neutrophil phenotype.
Collapse
Affiliation(s)
- Ricardo Grieshaber-Bouyer
- Division of Rheumatology, Department of Medicine V (Hematology, Oncology and Rheumatology), Heidelberg University Hospital, Heidelberg, Germany .,Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany.,Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), University of Heidelberg, Heidelberg, Germany.,Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tarik Exner
- Division of Rheumatology, Department of Medicine V (Hematology, Oncology and Rheumatology), Heidelberg University Hospital, Heidelberg, Germany.,Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nicolaj S Hackert
- Division of Rheumatology, Department of Medicine V (Hematology, Oncology and Rheumatology), Heidelberg University Hospital, Heidelberg, Germany.,Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix A Radtke
- Division of Rheumatology, Department of Medicine V (Hematology, Oncology and Rheumatology), Heidelberg University Hospital, Heidelberg, Germany.,Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Scott A Jelinsky
- Computational Systems Immunology, Worldwide Research & Development, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Olha Halyabar
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandra Wactor
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elham Karimizadeh
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph Brennan
- Computational Systems Immunology, Worldwide Research & Development, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Jorge Schettini
- Computational Systems Immunology, Worldwide Research & Development, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Helena Jonsson
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lauren A Henderson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carsten Müller-Tidow
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), University of Heidelberg, Heidelberg, Germany.,Department of Medicine V (Hematology Oncology Rheumatology), Heidelberg University Hospital, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V (Hematology, Oncology and Rheumatology), Heidelberg University Hospital, Heidelberg, Germany
| | - Guido Wabnitz
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Angela Hadjipanayis
- Computational Systems Immunology, Worldwide Research & Development, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Peter A Nigrovic
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA .,Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Cristescu R, Aurora-Garg D, Albright A, Xu L, Liu XQ, Loboda A, Lang L, Jin F, Rubin EH, Snyder A, Lunceford J. Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: a pan-tumor retrospective analysis of participants with advanced solid tumors. J Immunother Cancer 2022; 10:jitc-2021-003091. [PMID: 35101941 PMCID: PMC8804694 DOI: 10.1136/jitc-2021-003091] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Several studies have evaluated the relationship between tumor mutational burden (TMB) and outcomes of immune checkpoint inhibitors. In the phase II KEYNOTE-158 study of pembrolizumab monotherapy for previously treated recurrent or metastatic cancer, high TMB as assessed by the FoundationOne CDx was associated with an improved objective response rate (ORR). METHODS We retrospectively assessed the relationship between TMB and efficacy in participants with previously treated advanced solid tumors enrolled in 12 trials that evaluated pembrolizumab monotherapy, including 3 randomized trials that compared pembrolizumab with chemotherapy. TMB was assessed in formalin-fixed, paraffin-embedded pretreatment tumor samples by whole-exome sequencing. High TMB was defined as ≥175 mutations/exome. Microsatellite instability (MSI) phenotype was based on whole-exome sequencing results. Programmed death ligand 1 (PD-L1) expression was assessed by immunohistochemistry. The primary end point was ORR assessed per RECIST V.1.1 by independent central review. Other end points included progression-free survival (PFS) assessed per RECIST V.1.1 by independent central review and overall survival (OS). RESULTS Of the 2234 participants in the analysis, 1772 received pembrolizumab monotherapy and 462 received chemotherapy. Among the pembrolizumab-treated participants, ORR was 31.4% (95% CI 27.1 to 36.0) in the 433 participants with TMB ≥175 mutations/exome and 9.5% (95% CI 8.0 to 11.2) in the 1339 participants with TMB <175 mutations/exome. The association of TMB with ORR was observed regardless of PD-L1 expression and not driven by specific tumor types or participants with very high TMB or high MSI. In the 3 randomized controlled trials, TMB was associated with ORR (p≤0.016), PFS (p≤0.005), and OS (p≤0.029) of pembrolizumab but not of chemotherapy (p≥0.340, p≥0.643, and p≥0.174, respectively), and pembrolizumab improved efficacy versus chemotherapy in participants with TMB ≥175 mutations/exome. CONCLUSIONS TMB ≥175 mutations/exome is associated with clinically meaningful improvement in the efficacy of pembrolizumab monotherapy and improved outcomes for pembrolizumab versus chemotherapy across a wide range of previously treated advanced solid tumor types. These data suggest TMB has broad clinical utility irrespective of tumor type, PD-L1 expression, or MSI status and support its use as a predictive biomarker for pembrolizumab monotherapy in participants with previously treated advanced solid tumors.
Collapse
Affiliation(s)
| | | | | | - Lei Xu
- Merck & Co, Inc, Kenilworth, New Jersey, USA
| | | | | | - Lixin Lang
- Merck & Co, Inc, Kenilworth, New Jersey, USA
| | - Fan Jin
- Merck & Co, Inc, Kenilworth, New Jersey, USA
| | | | | | | |
Collapse
|
46
|
Drenkard C, Easley K, Bao G, Dunlop-Thomas C, Lim SS, Brady T. Cross-sectional study of the effects of self-efficacy on fatigue and pain interference in black women with systemic lupus erythematosus: the role of depression, age and education. Lupus Sci Med 2022; 9:9/1/e000566. [PMID: 35149578 PMCID: PMC8845307 DOI: 10.1136/lupus-2021-000566] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/06/2022] [Indexed: 11/17/2022]
Abstract
Objective While fatigue and pain are pervasive symptoms in SLE, self-efficacy can mitigate their intensity and impact on patients’ daily activity. We examined the relationships of these domains and their interactions with demographics and depression in black women with SLE. Methods This is a cross-sectional analysis of data collected among 699 black women with SLE. We used validated, self-reported measures of fatigue, pain interference, symptom self-efficacy, treatment self-efficacy and depression. Linear regression analyses were conducted to examine the relationships between each outcome (fatigue and pain interference) and each predictor (symptom self-efficacy and treatment self-efficacy), and the interaction of demographics and depression. Results We found inverse associations between fatigue and each of symptom self-efficacy (slope −0.556, p<0.001) and treatment self-efficacy (slope −0.282, p<0.001), as well as between pain interference and each of symptom self-efficacy (slope −0.394, p<0.001) and treatment self-efficacy (slope −0.152, p<0.001). After adjusting for confounders, symptom self-efficacy remained significantly associated with each outcome (adjusted slope −0.241 (p<0.001) and −0.103 (p=0.008) for fatigue and pain, respectively). The amount of decrease in fatigue and pain interference differed by depression severity (p<0.05 for the interaction of symptom self-efficacy and depression). The difference in fatigue by depression widened as symptom self-efficacy increased; the adjusted fatigue scores for moderate/severe depression compared with no depression were 6.8 and 8.7 points higher at mean and high symptom self-efficacy, respectively (p<0.001). Age and education significantly changed the relationship between outcomes and self-efficacy. Conclusions Symptom self-efficacy and treatment self-efficacy were inversely related to fatigue and pain interference in black women with SLE. Depression disproportionately increased the intensity of these outcomes. While older women with low symptom self-efficacy reported disproportionately higher pain interference, those with higher education and mean or high levels of symptom self-efficacy reported lower pain interference. These findings may help predict who might benefit most from self-efficacy-enhancing interventions.
Collapse
Affiliation(s)
- Cristina Drenkard
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, Georgia, USA .,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Kirk Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Gaobin Bao
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Charmayne Dunlop-Thomas
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - S Sam Lim
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Teresa Brady
- Clarity Consulting and Communications, Atlanta, Georgia, USA
| |
Collapse
|
47
|
Borst C, Symmank D, Drach M, Weninger W. Cutaneous signs and mechanisms of inflammasomopathies. Ann Rheum Dis 2022; 81:454-465. [PMID: 35039323 DOI: 10.1136/annrheumdis-2021-220977] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/20/2021] [Indexed: 11/03/2022]
Abstract
The emerging group of autoinflammatory diseases (AIDs) is caused by a dysregulation of the innate immune system while lacking the typical footprint of adaptive immunity. A prominent subgroup of AIDs are inflammasomopathies, which are characterised by periodic flares of cutaneous signs as well as systemic organ involvement and fever. The range of possible skin lesions is vast, ranging from urticarial, erysipelas-like and pustular rashes to erythematous patches, violaceous plaques and eventual necrosis and ulceration. This review provides a structured overview of the pathogenesis and the clinical picture with a focus on dermatological aspects of inflammasomopathies. Current treatment options for these conditions are also discussed.
Collapse
Affiliation(s)
- Carina Borst
- Department of Dermatology, Medical University of Vienna, Wien, Austria
| | - Dörte Symmank
- Department of Dermatology, Medical University of Vienna, Wien, Austria
| | - Mathias Drach
- Department of Dermatology, Medical University of Vienna, Wien, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Wien, Austria
| |
Collapse
|
48
|
Nagasawa Y, Misaki T, Ito S, Naka S, Wato K, Nomura R, Matsumoto-Nakano M, Nakano K. Title IgA Nephropathy and Oral Bacterial Species Related to Dental Caries and Periodontitis. Int J Mol Sci 2022; 23:725. [PMID: 35054910 PMCID: PMC8775524 DOI: 10.3390/ijms23020725] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
A relationship between IgA nephropathy (IgAN) and bacterial infection has been suspected. As IgAN is a chronic disease, bacteria that could cause chronic infection in oral areas might be pathogenetic bacteria candidates. Oral bacterial species related to dental caries and periodontitis should be candidates because these bacteria are well known to be pathogenic in chronic dental disease. Recently, several reports have indicated that collagen-binding protein (cnm)-(+) Streptococcs mutans is relate to the incidence of IgAN and the progression of IgAN. Among periodontal bacteria, Treponema denticola, Porphyromonas gingivalis and Campylobacte rectus were found to be related to the incidence of IgAN. These bacteria can cause IgAN-like histological findings in animal models. While the connection between oral bacterial infection, such as infection with S. mutans and periodontal bacteria, and the incidence of IgAN remains unclear, these bacterial infections might cause aberrantly glycosylated IgA1 in nasopharynx-associated lymphoid tissue, which has been reported to cause IgA deposition in mesangial areas in glomeruli, probably through the alteration of microRNAs related to the expression of glycosylation enzymes. The roles of other factors related to the incidence and progression of IgA, such as genes and cigarette smoking, can also be explained from the perspective of the relationship between these factors and oral bacteria. This review summarizes the relationship between IgAN and oral bacteria, such as cnm-(+) S. mutans and periodontal bacteria.
Collapse
Affiliation(s)
- Yasuyuki Nagasawa
- Department of General Internal Medicine, Hyogo College of Medicine, Nishinomiya 663-8501, Hyogo, Japan
| | - Taro Misaki
- Division of Nephrology, Seirei Hamamatsu General Hospital, Hamamatsu 430-8558, Shizuoka, Japan;
- Department of Nursing, Faculty of Nursing, Seirei Christopher University, Hamamatsu 433-8558, Shizuoka, Japan
| | - Seigo Ito
- Department of Internal Medicine, Japan Self-Defense Gifu Hospital, Kakamigahara 502-0817, Gifu, Japan;
| | - Shuhei Naka
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Okayama, Japan; (S.N.); (M.M.-N.)
| | - Kaoruko Wato
- Department of Pediatric Dentistry, Division of Oral Infection and Disease Control, Osaka University Graduate School of Dentistry, Suita 565-0871, Osaka, Japan; (K.W.); (R.N.); (K.N.)
| | - Ryota Nomura
- Department of Pediatric Dentistry, Division of Oral Infection and Disease Control, Osaka University Graduate School of Dentistry, Suita 565-0871, Osaka, Japan; (K.W.); (R.N.); (K.N.)
| | - Michiyo Matsumoto-Nakano
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Okayama, Japan; (S.N.); (M.M.-N.)
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Division of Oral Infection and Disease Control, Osaka University Graduate School of Dentistry, Suita 565-0871, Osaka, Japan; (K.W.); (R.N.); (K.N.)
| |
Collapse
|
49
|
Hofer S, Hofstätter N, Punz B, Hasenkopf I, Johnson L, Himly M. Immunotoxicity of nanomaterials in health and disease: Current challenges and emerging approaches for identifying immune modifiers in susceptible populations. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1804. [PMID: 36416020 PMCID: PMC9787548 DOI: 10.1002/wnan.1804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 11/24/2022]
Abstract
Nanosafety assessment has experienced an intense era of research during the past decades driven by a vivid interest of regulators, industry, and society. Toxicological assays based on in vitro cellular models have undergone an evolution from experimentation using nanoparticulate systems on singular epithelial cell models to employing advanced complex models more realistically mimicking the respective body barriers for analyzing their capacity to alter the immune state of exposed individuals. During this phase, a number of lessons were learned. We have thus arrived at a state where the next chapters have to be opened, pursuing the following objectives: (1) to elucidate underlying mechanisms, (2) to address effects on vulnerable groups, (3) to test material mixtures, and (4) to use realistic doses on (5) sophisticated models. Moreover, data reproducibility has become a significant demand. In this context, we studied the emerging concept of adverse outcome pathways (AOPs) from the perspective of immune activation and modulation resulting in pro-inflammatory versus tolerogenic responses. When considering the interaction of nanomaterials with biological systems, protein corona formation represents the relevant molecular initiating event (e.g., by potential alterations of nanomaterial-adsorbed proteins). Using this as an example, we illustrate how integrated experimental-computational workflows combining in vitro assays with in silico models aid in data enrichment and upon comprehensive ontology-annotated (meta)data upload to online repositories assure FAIRness (Findability, Accessibility, Interoperability, Reusability). Such digital twinning may, in future, assist in early-stage decision-making during therapeutic development, and hence, promote safe-by-design innovation in nanomedicine. Moreover, it may, in combination with in silico-based exposure-relevant dose-finding, serve for risk monitoring in particularly loaded areas, for example, workplaces, taking into account pre-existing health conditions. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Sabine Hofer
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Norbert Hofstätter
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Benjamin Punz
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Ingrid Hasenkopf
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Litty Johnson
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| | - Martin Himly
- Division of Allergy & Immunology, Department of Biosciences & Medical BiologyParis Lodron University of SalzburgSalzburgAustria
| |
Collapse
|
50
|
Hecht M, Eckstein M, Rutzner S, von der Grün J, Illmer T, Klautke G, Laban S, Hautmann MG, Brunner TB, Tamaskovics B, Hinke A, Zhou JG, Frey B, Donaubauer AJ, Becker I, Semrau S, Hartmann A, Balermpas P, Budach W, Gaipl US, Iro H, Gostian AO, Fietkau R. Induction chemoimmunotherapy followed by CD8+ immune cell-based patient selection for chemotherapy-free radioimmunotherapy in locally advanced head and neck cancer. J Immunother Cancer 2022; 10:e003747. [PMID: 35078923 PMCID: PMC8796267 DOI: 10.1136/jitc-2021-003747] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2021] [Indexed: 01/05/2023] Open
Abstract
PURPOSE The first aim of the trial is to study feasibility of combined programmed death protein ligand 1/cytotoxic T-lymphocyte-associated protein 4 inhibition concomitant to radiotherapy. In addition, efficacy of the entire treatment scheme consisting of induction chemoimmunotherapy followed by chemotherapy-free radioimmunotherapy (RIT) after intratumoral CD8 +immune cell-based patient selection will be analyzed. METHODS Patients with stage III-IVB head and neck squamous cell carcinoma were eligible for this multicenter phase II trial. Treatment consisted of a single cycle of cisplatin 30 mg/m² days 1-3, docetaxel 75 mg/m² day 1, durvalumab 1500 mg fix dose day 5 and tremelimumab 75 mg fix dose day 5. Patients with increased intratumoral CD8 +immune cell density or pathological complete response (pCR) in the rebiopsy entered RIT up to a total dose of 70 Gy. Patients received further three cycles of durvalumab/tremelimumab followed by eight cycles of durvalumab mono (every 4 weeks). The intended treatment for patients not meeting these criteria was standard radiochemotherapy outside the trial. Primary endpoint was a feasibility rate of patients entering RIT to receive treatment until at least cycle 6 of immunotherapy of ≥80%. RESULTS Between September 2018 and May 2020, 80 patients were enrolled (one excluded). Out of these, 23 patients had human papilloma virus (HPV)-positive oropharyngeal cancer. Median follow-up was 17.2 months. After induction chemoimmunotherapy 41 patients had pCR and 31 had increased intratumoral CD8 +immune cells. Of 60 patients entering RIT (primary endpoint cohort), 10 experienced imiting toxic (mainly hepatitis) and four discontinued for other reasons, resulting in a feasibility rate of 82%. The RIT cohort (n=60) had a progression-free survival (PFS) rate at one and 2 years of 78% and 72%, respectively, and an overall survival rate at one and 2 years of 90% and 84%, respectively. Patients with HPV-positive oropharyngeal cancers had greater benefit from RIT with a 2-year PFS rate of 94% compared with 64% for HPV-negative oropharyngeal cancers and other locations. In the entire study cohort (n=79) the 2-year PFS rate was 68% (91% for HPV-positive oropharynx vs 59% for others). Toxicity grade 3-4 mainly consisted of dysphagia (53%), leukopenia (52%) and infections (32%). CONCLUSIONS The trial met the primary endpoint feasibility of RIT. Induction chemo-immunotherapy followed by chemotherapy-free RIT after intratumoral CD8 +immune cell-based patient selection has promising PFS. TRIAL REGISTRATION NUMBER The trial was registered with ClinicalTrials.gov (identifier: NCT03426657). The trial was conducted as investigator-sponsored trial (IST).
Collapse
Affiliation(s)
- Markus Hecht
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Markus Eckstein
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sandra Rutzner
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Jens von der Grün
- Department of Radiation Oncology, University Hospital Frankfurt, Goethe-Universitat Frankfurt am Main, Frankfurt am Main, Germany
| | - Thomas Illmer
- Private Praxis Oncology, Arnoldstraße, Dresden, Germany
| | - Gunther Klautke
- Department of Radiation Oncology, Chemnitz Hospital, Chemnitz, Germany
| | - Simon Laban
- Department of Otolaryngology - Head & Neck Surgery, Universität Ulm, Ulm, Germany
| | - Matthias G Hautmann
- Department of Radiotherapy, University Hospital Regensburg, Universität Regensburg, Regensburg, Germany
| | - Thomas B Brunner
- Department of Radiation Oncology, University Hospital Magdeburg, Otto von Guericke Universität Magdeburg, Magdeburg, Germany
| | - Bálint Tamaskovics
- Department of Radiation Oncology, University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Dusseldorf, Germany
| | - Axel Hinke
- Clinical Cancer Research Consulting (CCRC), Düsseldorf, Germany
| | - Jian-Guo Zhou
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Anna-Jasmina Donaubauer
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Ina Becker
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Sabine Semrau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Arndt Hartmann
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Panagiotis Balermpas
- Department of Radiation Oncology, University Hospital Frankfurt, Goethe-Universitat Frankfurt am Main, Frankfurt am Main, Germany
| | - Wilfried Budach
- Department of Radiation Oncology, University Hospital Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Dusseldorf, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Heinrich Iro
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
- Department of Otolaryngology - Head & Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Antoniu-Oreste Gostian
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
- Department of Otolaryngology - Head & Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| |
Collapse
|