1
|
Zaroon, Aslam S, Hafsa, Mustafa U, Fatima S, Bashir H. Interleukin in Immune-Mediated Diseases: An Updated Review. Mol Biotechnol 2024:10.1007/s12033-024-01347-8. [PMID: 39715931 DOI: 10.1007/s12033-024-01347-8] [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: 08/20/2024] [Accepted: 11/29/2024] [Indexed: 12/25/2024]
Abstract
The immune system comprises various regulators and effectors that elicit immune responses against various attacks on the body. The pathogenesis of autoimmune diseases is derived from the deregulated expression of cytokines, the major regulators of the immune system. Among cytokines, interleukins have a major influence on immune-mediated diseases. These interleukins initiate the immune response against healthy and normal cells of the body, resulting in immune-mediated disease. The major interleukins in this respect are IL-1, IL-3, IL-4, IL-6, IL-10 and IL-12 which cause immune responses such as excessive inflammation, loss of immune tolerance, altered T-cell differentiation, immune suppression dysfunction, and inflammatory cell recruitment. Systemic Lupus Erythematosus (SLE) is an autoimmune illness characterized by dysregulation of interleukins. These immune responses are the signs of diseases such as rheumatoid arthritis, inflammatory bowel disease, psoriasis, type I diabetes, and multiple sclerosis.
Collapse
Affiliation(s)
- Zaroon
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Shakira Aslam
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hafsa
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Usama Mustafa
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Sana Fatima
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hamid Bashir
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
2
|
Chen H, Liu L, Wang Y, Hong L, Pan J, Yu X, Dai H. Managing Cardiovascular Risk in Patients with Autoimmune Diseases: Insights from a Nutritional Perspective. Curr Nutr Rep 2024; 13:718-728. [PMID: 39078574 DOI: 10.1007/s13668-024-00563-7] [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] [Accepted: 07/19/2024] [Indexed: 07/31/2024]
Abstract
PURPOSE OF REVIEW Autoimmune diseases manifest as an immune system response directed against endogenous antigens, exerting a significant influence on a substantial portion of the population. Notably, a leading contributor to morbidity and mortality in this context is cardiovascular disease (CVD). Intriguingly, individuals with autoimmune disorders exhibit a heightened prevalence of CVD compared to the general population. The meticulous management of CV risk factors assumes paramount importance, given the current absence of a standardized solution to this perplexity. This review endeavors to address this challenge from a nutritional perspective. RECENT FINDINGS Emerging evidence suggests that inflammation, a common thread in autoimmune diseases, also plays a pivotal role in the pathogenesis of CVD. Nutritional interventions aimed at reducing inflammation have shown promise in mitigating cardiovascular risk. The integration of nutritional strategies into the management plans for patients with autoimmune diseases offers a holistic approach to reducing cardiovascular risk. While conventional pharmacological treatments remain foundational, the addition of targeted dietary interventions can provide a complementary pathway to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Huimin Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Lu Liu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Yi Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Liqiong Hong
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Jiahui Pan
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Xiongkai Yu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Haijiang Dai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China.
| |
Collapse
|
3
|
Konozy EH, Osman MEM. From inflammation to immune regulation: The dual nature of dietary lectins in health and disease. Heliyon 2024; 10:e39471. [PMID: 39502251 PMCID: PMC11535980 DOI: 10.1016/j.heliyon.2024.e39471] [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: 06/24/2023] [Revised: 09/24/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
Beans, vegetables, fruits, and mushrooms offer a delightful array of fragrances and an abundance of nutrients, including essential vitamins, minerals, protein rich in vital amino acids, and omega-3 fatty acids. However, they may also contain lectins, carbohydrate-binding proteins with potential health risks. While some lectins exhibit stability and resistance to digestion, posing threats to gastrointestinal integrity and immune function, others, such as those from butterfly peas and pink bauhinia, show immunomodulatory properties that could bolster immune responses. While some lectins, such as phytohemagglutinin, have been associated with inflammatory responses and autoimmune disorders, others, such as wheat lectin, have shown potential benefits in nutrient absorption. Additionally, mushroom lectins, while generally nontoxic, exhibit immunomodulatory properties with implications for immune health. Despite their potential benefits, challenges remain in understanding lectin dosages, administration routes, and mechanisms of action. Further research is needed to elucidate the intricate roles of dietary lectins in immune function and autoimmune disorders. This review surveys the immunomodulatory effects of dietary lectins from plants and mushrooms, shedding light on their mechanisms of action. From inflammation modulation to potential autoimmune implications, the diverse roles of dietary lectins have been explored, highlighting avenues for future investigations and therapeutic exploration.
Collapse
Affiliation(s)
- Emadeldin H.E. Konozy
- Biotechnology Department, Africa City of Technology, Khartoum, Sudan
- Pharmaceutical Research and Development Centre, Faculty of Pharmacy, Karary University Omdurman, Khartoum State, Sudan
- Biomedical and Clinical Research Centre (BCRC), College of Health and Allied Sciences (CoHAS), University of Cape Coast, Cape Coast, Ghana
| | | |
Collapse
|
4
|
Schneider KM, Kummen M, Trivedi PJ, Hov JR. Role of microbiome in autoimmune liver diseases. Hepatology 2024; 80:965-987. [PMID: 37369002 PMCID: PMC11407779 DOI: 10.1097/hep.0000000000000506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/25/2023] [Indexed: 06/29/2023]
Abstract
The microbiome plays a crucial role in integrating environmental influences into host physiology, potentially linking it to autoimmune liver diseases, such as autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis. All autoimmune liver diseases are associated with reduced diversity of the gut microbiome and altered abundance of certain bacteria. However, the relationship between the microbiome and liver diseases is bidirectional and varies over the course of the disease. This makes it challenging to dissect whether such changes in the microbiome are initiating or driving factors in autoimmune liver diseases, secondary consequences of disease and/or pharmacological intervention, or alterations that modify the clinical course that patients experience. Potential mechanisms include the presence of pathobionts, disease-modifying microbial metabolites, and more nonspecific reduced gut barrier function, and it is highly likely that the effect of these change during the progression of the disease. Recurrent disease after liver transplantation is a major clinical challenge and a common denominator in these conditions, which could also represent a window to disease mechanisms of the gut-liver axis. Herein, we propose future research priorities, which should involve clinical trials, extensive molecular phenotyping at high resolution, and experimental studies in model systems. Overall, autoimmune liver diseases are characterized by an altered microbiome, and interventions targeting these changes hold promise for improving clinical care based on the emerging field of microbiota medicine.
Collapse
Affiliation(s)
| | - Martin Kummen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Palak J. Trivedi
- National Institute for Health and Care Research Birmingham Biomedical Research Centre, Centre for Liver and Gastroenterology Research, University of Birmingham, UK
- Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, UK
- Institute of Applied Health Research, University of Birmingham, UK
| | - Johannes R. Hov
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
5
|
Trivedi PJ, Hirschfield GM, Adams DH, Vierling JM. Immunopathogenesis of Primary Biliary Cholangitis, Primary Sclerosing Cholangitis and Autoimmune Hepatitis: Themes and Concepts. Gastroenterology 2024; 166:995-1019. [PMID: 38342195 DOI: 10.1053/j.gastro.2024.01.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 01/21/2024] [Accepted: 01/28/2024] [Indexed: 02/13/2024]
Abstract
Autoimmune liver diseases include primary biliary cholangitis, primary sclerosing cholangitis, and autoimmune hepatitis, a family of chronic immune-mediated disorders that target hepatocytes and cholangiocytes. Treatments remain nonspecific, variably effective, and noncurative, and the need for liver transplantation is disproportionate to their rarity. Development of effective therapies requires better knowledge of pathogenic mechanisms, including the roles of genetic risk, and how the environment and gut dysbiosis cause immune cell dysfunction and aberrant bile acid signaling. This review summarizes key etiologic and pathogenic concepts and themes relevant for clinical practice and how such learning can guide the development of new therapies for people living with autoimmune liver diseases.
Collapse
Affiliation(s)
- Palak J Trivedi
- National Institute for Health Research Birmingham Biomedical Research Centre, Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom; Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom; Institute of Translational Medicine, University of Birmingham, Birmingham, United Kingdom.
| | - Gideon M Hirschfield
- Division of Gastroenterology and Hepatology, Toronto Centre for Liver Disease, University of Toronto, Toronto, Ontario, Canada
| | - David H Adams
- National Institute for Health Research Birmingham Biomedical Research Centre, Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom; Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
| | - John M Vierling
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Division of Abdominal Transplantation, Department of Surgery, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
6
|
Chen Y, Feng S, Li Y, Zhang C, Chao G, Zhang S. Gut microbiota and intestinal immunity-A crosstalk in irritable bowel syndrome. Immunology 2024; 172:1-20. [PMID: 38174581 DOI: 10.1111/imm.13749] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Irritable bowel syndrome (IBS), one of the most prevalent functional gastrointestinal disorders, is characterized by recurrent abdominal pain and abnormal defecation habits, resulting in a severe healthcare burden worldwide. The pathophysiological mechanisms of IBS are multi-factorially involved, including food antigens, visceral hypersensitivity reactions, and the brain-gut axis. Numerous studies have found that gut microbiota and intestinal mucosal immunity play an important role in the development of IBS in crosstalk with multiple mechanisms. Therefore, based on existing evidence, this paper elaborates that the damage and activation of intestinal mucosal immunity and the disturbance of gut microbiota are closely related to the progression of IBS. Combined with the application prospect, it also provides references for further in-depth exploration and clinical practice.
Collapse
Affiliation(s)
- Yuxuan Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyan Feng
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Li
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Zhang
- Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, China
| | - Guanqun Chao
- Department of General Practice, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, China
| | - Shuo Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Lerner A, Benzvi C, Vojdani A. Cross-reactivity and sequence similarity between microbial transglutaminase and human tissue antigens. Sci Rep 2023; 13:17526. [PMID: 37845267 PMCID: PMC10579360 DOI: 10.1038/s41598-023-44452-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023] Open
Abstract
Microbial transglutaminase (mTG) is a bacterial survival factor, frequently used as a food additive to glue processed nutrients. As a result, new immunogenic epitopes are generated that might drive autoimmunity. Presently, its contribution to autoimmunity through epitope similarity and cross-reactivity was investigated. Emboss Matcher was used to perform sequence alignment between mTG and various antigens implicated in many autoimmune diseases. Monoclonal and polyclonal antibodies made specifically against mTG were applied to 77 different human tissue antigens using ELISA. Six antigens were detected to share significant homology with mTG immunogenic sequences, representing major targets of common autoimmune conditions. Polyclonal antibody to mTG reacted significantly with 17 out of 77 tissue antigens. This reaction was most pronounced with mitochondrial M2, ANA, and extractable nuclear antigens. The results indicate that sequence similarity and cross-reactivity between mTG and various tissue antigens are possible, supporting the relationship between mTG and the development of autoimmune disorders 150W.
Collapse
Affiliation(s)
- Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel.
- Ariel University, Ariel, Israel.
| | - Carina Benzvi
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| | | |
Collapse
|
8
|
Lampousi AM, Carlsson S, Löfvenborg JE, Cabrera-Castro N, Chirlaque MD, Fagherazzi G, Franks PW, Hampe CS, Jakszyn P, Koulman A, Kyrø C, Moreno-Iribas C, Nilsson PM, Panico S, Papier K, van der Schouw YT, Schulze MB, Weiderpass E, Zamora-Ros R, Forouhi NG, Sharp SJ, Rolandsson O, Wareham NJ. Interaction between plasma phospholipid odd-chain fatty acids and GAD65 autoantibodies on the incidence of adult-onset diabetes: the EPIC-InterAct case-cohort study. Diabetologia 2023; 66:1460-1471. [PMID: 37301794 PMCID: PMC10317878 DOI: 10.1007/s00125-023-05948-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/25/2023] [Indexed: 06/12/2023]
Abstract
AIMS/HYPOTHESIS Islet autoimmunity may progress to adult-onset diabetes. We investigated whether circulating odd-chain fatty acids (OCFA) 15:0 and 17:0, which are inversely associated with type 2 diabetes, interact with autoantibodies against GAD65 (GAD65Ab) on the incidence of adult-onset diabetes. METHODS We used the European EPIC-InterAct case-cohort study including 11,124 incident adult-onset diabetes cases and a subcohort of 14,866 randomly selected individuals. Adjusted Prentice-weighted Cox regression estimated HRs and 95% CIs of diabetes in relation to 1 SD lower plasma phospholipid 15:0 and/or 17:0 concentrations or their main contributor, dairy intake, among GAD65Ab-negative and -positive individuals. Interactions between tertiles of OCFA and GAD65Ab status were estimated by proportion attributable to interaction (AP). RESULTS Low concentrations of OCFA, particularly 17:0, were associated with a higher incidence of adult-onset diabetes in both GAD65Ab-negative (HR 1.55 [95% CI 1.48, 1.64]) and GAD65Ab-positive (HR 1.69 [95% CI 1.34, 2.13]) individuals. The combination of low 17:0 and high GAD65Ab positivity vs high 17:0 and GAD65Ab negativity conferred an HR of 7.51 (95% CI 4.83, 11.69), with evidence of additive interaction (AP 0.25 [95% CI 0.05, 0.45]). Low dairy intake was not associated with diabetes incidence in either GAD65Ab-negative (HR 0.98 [95% CI 0.94, 1.02]) or GAD65Ab-positive individuals (HR 0.97 [95% CI 0.79, 1.18]). CONCLUSIONS/INTERPRETATION Low plasma phospholipid 17:0 concentrations may promote the progression from GAD65Ab positivity to adult-onset diabetes.
Collapse
Affiliation(s)
- Anna-Maria Lampousi
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Sofia Carlsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josefin E Löfvenborg
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Risk and Benefit Assessment, Swedish Food Agency, Uppsala, Sweden
| | | | - María-Dolores Chirlaque
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Health and Social Sciences, Murcia University, Murcia, Spain
| | - Guy Fagherazzi
- Deep Digital Phenotyping Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Paul W Franks
- Department of Clinical Sciences, Clinical Research Center, Skåne University Hospital, Lund University, Malmö, Sweden
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Umeå, Sweden
| | - Christiane S Hampe
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Albert Koulman
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- National Institute for Health Research Biomedical Research Centre Core Nutritional Biomarker Laboratory, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Cecilie Kyrø
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Conchi Moreno-Iribas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Peter M Nilsson
- Department of Clinical Sciences, Clinical Research Center, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Keren Papier
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Nita G Forouhi
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Stephen J Sharp
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Olov Rolandsson
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Umeå, Sweden
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
9
|
Li Q, Meng LB, Chen LJ, Shi X, Tu L, Zhou Q, Yu JL, Liao X, Zeng Y, Yuan QY. The role of the microbiota-gut-brain axis and intestinal microbiome dysregulation in Parkinson's disease. Front Neurol 2023; 14:1185375. [PMID: 37305758 PMCID: PMC10249504 DOI: 10.3389/fneur.2023.1185375] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/24/2023] [Indexed: 06/13/2023] Open
Abstract
Parkinson's disease (PD) is a complex progressive neurodegenerative disease associated with aging. Its main pathological feature is the degeneration and loss of dopaminergic neurons related to the misfolding and aggregation of α-synuclein. The pathogenesis of PD has not yet been fully elucidated, and its occurrence and development process are closely related to the microbiota-gut-brain axis. Dysregulation of intestinal microbiota may promote the damage of the intestinal epithelial barrier, intestinal inflammation, and the upward diffusion of phosphorylated α-synuclein from the enteric nervous system (ENS) to the brain in susceptible individuals and further lead to gastrointestinal dysfunction, neuroinflammation, and neurodegeneration of the central nervous system (CNS) through the disordered microbiota-gut-brain axis. The present review aimed to summarize recent advancements in studies focusing on the role of the microbiota-gut-brain axis in the pathogenesis of PD, especially the mechanism of intestinal microbiome dysregulation, intestinal inflammation, and gastrointestinal dysfunction in PD. Maintaining or restoring homeostasis in the gut microenvironment by targeting the gut microbiome may provide future direction for the development of new biomarkers for early diagnosis of PD and therapeutic strategies to slow disease progression.
Collapse
Affiliation(s)
- Qing Li
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Ling-bing Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Li-jun Chen
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Xia Shi
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Ling Tu
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Qi Zhou
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Jin-long Yu
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Xin Liao
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Yuan Zeng
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| | - Qiao-ying Yuan
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), The First Affiliated Hospital of PLA Army Medical University, Chongqing, China
| |
Collapse
|
10
|
Ageeli RY, Sharma S, Puppa M, Bloomer RJ, Buddington RK, van der Merwe M. Fasting Protocols Do Not Improve Intestinal Architecture and Immune Parameters in C57BL/6 Male Mice Fed a High Fat Diet. MEDICINES (BASEL, SWITZERLAND) 2023; 10:18. [PMID: 36827218 PMCID: PMC9961949 DOI: 10.3390/medicines10020018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND The intestinal ecosystem, including epithelium, immune cells, and microbiota, are influenced by diet and timing of food consumption. The purpose of this study was to evaluate various dietary protocols after ad libitum high fat diet (HFD) consumption on intestinal morphology and mucosal immunity. METHODS C57BL/6 male mice were fed a 45% high fat diet (HFD) for 6 weeks and then randomized to the following protocols; (1) chow, (2) a purified high fiber diet known as the Daniel Fast (DF), HFD consumed (3) ad libitum or in a restricted manner; (4) caloric-restricted, (5) time-restricted (six hours of fasting in each 24 h), or (6) alternate-day fasting (24 h fasting every other day). Intestinal morphology and gut-associated immune parameters were investigated after 2 months on respective protocols. RESULTS Consuming a HFD resulted in shortening of the intestine and reduction in villi and crypt size. Fasting, while consuming the HFD, did not restore these parameters to the extent seen with the chow and DF diet. Goblet cell number and regulatory T cells had improved recovery with high fiber diets, not seen with the HFD irrespective of fasting. CONCLUSION Nutritional content is a critical determinant of intestinal parameters associated with gut health.
Collapse
Affiliation(s)
| | | | | | | | | | - Marie van der Merwe
- College of Health Sciences, The University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|
11
|
Bakulin IG, Avalueva EB, Semenova EA, Оrеshkо LS, Serkova MY, Sitkin SI. Prospects for the treatment of gluten-associated diseases: on our daily bread, celiac disease, gluten proteins and more…. ALMANAC OF CLINICAL MEDICINE 2023; 50:367-376. [DOI: 10.18786/2072-0505-2022-50-053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Food safety all over the world is largely dependent on production of grains that are cultivated in 60% of agricultural lands. Wheat is the main food for millions of people and one of the three most commonly cultivated grain cultures worldwide, along with corn and rice. Modern wheat is a product of gene engineering interventions aimed at increased productivity, yields, nutrient quota, and storage time, as well as immunogenic properties. However, the consumption of gluten, a proline and glutamine-rich wheat, rye and barley protein, triggers gluten-dependent disorders, such as celiac disease, wheat allergy, baker's asthma and wheat-dependent exercise-induced anaphylaxis. This group of disorders are curable provided the correct diagnosis has been made and strict lifelong gluten-free diet is implemented. Continuous patient's adherence to the gluten-free diet is associated with a number of medical and paramedical challenges, and the adherence level of the most compliant patients does not exceed 80%. The paper discuss other treatment strategies to improve the nutrition of people with gluten-sensitive disorders, in particular, the reduction grain gluten content, gluten sequestration in the gut before its digestion, prevention of gluten absorption and subsequent immune cell activation, and administration of tissue transglutaminase 2 inhibitors.
Collapse
|
12
|
Vojdani A, Vojdani E, Melgar AL, Redd J. Reaction of SARS-CoV-2 antibodies with other pathogens, vaccines, and food antigens. Front Immunol 2022; 13:1003094. [PMID: 36211404 PMCID: PMC9537454 DOI: 10.3389/fimmu.2022.1003094] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
It has been shown that SARS-CoV-2 shares homology and cross-reacts with vaccines, other viruses, common bacteria and many human tissues. We were inspired by these findings, firstly, to investigate the reaction of SARS-CoV-2 monoclonal antibody with different pathogens and vaccines, particularly DTaP. Additionally, since our earlier studies have shown immune reactivity by antibodies made against pathogens and autoantigens towards different food antigens, we also studied cross-reaction between SARS-CoV-2 and common foods. For this, we reacted monoclonal and polyclonal antibodies against SARS-CoV-2 spike protein and nucleoprotein with 15 different bacterial and viral antigens and 2 different vaccines, BCG and DTaP, as well as with 180 different food peptides and proteins. The strongest reaction by SARS-CoV-2 antibodies were with DTaP vaccine antigen, E. faecalis, roasted almond, broccoli, soy, cashew, α+β casein and milk, pork, rice endochitinase, pineapple bromelain, and lentil lectin. Because the immune system tends to form immune responses towards the original version of an antigen that it has encountered, this cross-reactivity may have its advantages with regards to immunity against SARS-CoV-2, where the SARS-CoV-2 virus may elicit a “remembered” immune response because of its structural similarity to a pathogen or food antigen to which the immune system was previously exposed. Our findings indicate that cross-reactivity elicited by DTaP vaccines in combination with common herpesviruses, bacteria that are part of our normal flora such as E. faecalis, and foods that we consume on a daily basis should be investigated for possible cross-protection against COVID-19. Additional experiments would be needed to clarify whether or not this cross-protection is due to cross-reactive antibodies or long-term memory T and B cells in the blood.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab, Los Angeles, CA, United States
- Cyrex Laboratories, Limited Liability Company (LLC), Phoenix, AZ, United States
- *Correspondence: Aristo Vojdani,
| | | | | | - Joshua Redd
- RedRiver Health and Wellness, South Jordan, UT, United States
| |
Collapse
|
13
|
Marrugo Padilla A, Rizzo G, Smaldini PL, Vaccaro J, Méndez Cuadro D, Rodríguez Cavallo E, Docena GH. Carbonylation induced by antibiotic and pesticide residues on casein increases its IgE binding and allergenicity. Free Radic Res 2022; 56:28-39. [DOI: 10.1080/10715762.2022.2032020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Albeiro Marrugo Padilla
- Analytical Chemistry and Biomedicine Group. Campus of Zaragocilla ancient building Cread. University of Cartagena. Cartagena-Colombia.
| | - Gastón Rizzo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| | - Paola L. Smaldini
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| | - Julián Vaccaro
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| | - Darío Méndez Cuadro
- Analytical Chemistry and Biomedicine Group. Campus of Zaragocilla ancient building Cread. University of Cartagena. Cartagena-Colombia.
| | - Erika Rodríguez Cavallo
- Analytical Chemistry and Biomedicine Group. Campus of Zaragocilla ancient building Cread. University of Cartagena. Cartagena-Colombia.
| | - Guillermo H. Docena
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| |
Collapse
|
14
|
Knippenberg A, Robinson GA, Wincup C, Ciurtin C, Jury EC, Kalea AZ. Plant-based dietary changes may improve symptoms in patients with systemic lupus erythematosus. Lupus 2022; 31:65-76. [PMID: 34978516 PMCID: PMC8793314 DOI: 10.1177/09612033211063795] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Previous studies have reported that patients affected by systemic lupus erythematosus (SLE) are interested in using diet to treat fatigue, cardiovascular disease and other symptoms. However, to date, there is insufficient information regarding the ways for patients to modify their diet to improve SLE symptoms. We investigated the relationship between the eating patterns of SLE patients and their self-reported disease symptoms and general aspects of health. METHODS A UK-based, online survey was developed, in which patients with SLE were asked about their attitudes and experiences regarding their SLE symptoms and diet. RESULTS The majority (>80%) of respondents that undertook new eating patterns with increased vegetable intake and/or decreased intake of processed food, sugar, gluten, dairy and carbohydrates reported benefiting from their dietary change. Symptom severity ratings after these dietary changes were significantly lower than before (21.3% decrease, p<0.0001). The greatest decreases in symptom severity were provided by low/no dairy (27.1% decrease), low/no processed foods (26.6% decrease) and vegan (26% decrease) eating patterns (p<0.0001). Weight loss, fatigue, joint/muscle pain and mood were the most cited symptoms that improved with dietary change. CONCLUSION SLE patients who changed their eating patterns to incorporate more plant-based foods while limiting processed foods and animal products reported improvements in their disease symptoms. Thus, our findings show promises in using nutrition interventions for the management of SLE symptoms, setting the scene for future clinical trials in this area. Randomised studies are needed to further test whether certain dietary changes are effective for improving specific symptoms of SLE.
Collapse
Affiliation(s)
| | - George A Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Chris Wincup
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Anastasia Z Kalea
- Division of Medicine, University College London, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| |
Collapse
|
15
|
The Role of Exposomes in the Pathophysiology of Autoimmune Diseases I: Toxic Chemicals and Food. PATHOPHYSIOLOGY 2021; 28:513-543. [PMID: 35366249 PMCID: PMC8830458 DOI: 10.3390/pathophysiology28040034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/11/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Autoimmune diseases affect 5–9% of the world’s population. It is now known that genetics play a relatively small part in the pathophysiology of autoimmune disorders in general, and that environmental factors have a greater role. In this review, we examine the role of the exposome, an individual’s lifetime exposure to external and internal factors, in the pathophysiology of autoimmune diseases. The most common of these environmental factors are toxic chemicals, food/diet, and infections. Toxic chemicals are in our food, drink, common products, the air, and even the land we walk on. Toxic chemicals can directly damage self-tissue and cause the release of autoantigens, or can bind to human tissue antigens and form neoantigens, which can provoke autoimmune response leading to autoimmunity. Other types of autoimmune responses can also be induced by toxic chemicals through various effects at the cellular and biochemical levels. The food we eat every day commonly has colorants, preservatives, or packaging-related chemical contamination. The food itself may be antigenic for susceptible individuals. The most common mechanism for food-related autoimmunity is molecular mimicry, in which the food’s molecular structure bears a similarity with the structure of one or more self-tissues. The solution is to detect the trigger, remove it from the environment or diet, then repair the damage to the individual’s body and health.
Collapse
|
16
|
Lampousi AM, Carlsson S, Löfvenborg JE. Dietary factors and risk of islet autoimmunity and type 1 diabetes: a systematic review and meta-analysis. EBioMedicine 2021; 72:103633. [PMID: 34656932 PMCID: PMC8523874 DOI: 10.1016/j.ebiom.2021.103633] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Background Numerous dietary components have been linked to the development of islet autoimmunity (IA) and type 1 diabetes (T1D); however, no associations are firmly established. This systematic review and meta-analysis aimed to synthesize current knowledge on diet and incidence of IA and T1D. Methods Literature search was performed in Medline, Embase, and Cochrane Library, from inception until October 2020. Eligible studies had IA or T1D as outcome; any dietary exposure; case-control, cohort, or randomized controlled trial design; and hazard, risk, or odds ratios as measures of association. Summary relative risks (RR) and 95% confidence intervals (CI) were estimated with random-effects models. Certainty of evidence was assessed with GRADE. PROSPERO registration number: CRD42020212505. Findings Among 5935 identified records, 96 were eligible, and pooled estimates could be produced for 26 dietary factors. Evidence with moderate/high certainty indicated lower risk of T1D in relation to longer (≥6-12 vs <6-12 months, RR: 0⋅39, CI: 0⋅26-0⋅58, I2=43%) and exclusive (≥2-3 vs <2-3 months, RR: 0⋅68, CI: 0⋅58-0⋅80, I2=0%) breastfeeding, later introduction to gluten (3-6 vs <3-5 months, RR: 0⋅36, CI: 0⋅17-0⋅75, I2=0%), cow's milk (≥2-3 vs <2-3 months, RR: 0⋅69, CI: 0⋅59-0⋅81, I2=0%), and fruit (4-6 vs <4-5 months, RR: 0⋅47, CI: 0⋅25-0⋅86, I2=0%). Higher childhood intake of cow's milk was associated with increased risk of both IA (per 2-3 portions/day, RR: 1⋅25, CI: 1⋅06-1⋅47, I2=0%) and T1D (≥2-3 vs <2-3 glasses/day, RR: 1⋅81, CI: 1⋅12-2⋅91, I2=31%). For the remaining dietary factors investigated, there was no association, or the evidence was of low certainty. Interpretation This study suggests that breastfeeding and late introduction of gluten, fruit, and cow's milk may reduce the risk of T1D, whereas high childhood cow's milk intake may increase it. Funding Swedish Research Council, Swedish Research Council for Health, Working Life and Welfare (FORTE), Novo Nordisk Foundation, and Swedish Diabetes Foundation.
Collapse
Affiliation(s)
- Anna-Maria Lampousi
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Sofia Carlsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
17
|
Al-Ayadhi L, Zayed N, Bhat RS, Moubayed NMS, Al-Muammar MN, El-Ansary A. The use of biomarkers associated with leaky gut as a diagnostic tool for early intervention in autism spectrum disorder: a systematic review. Gut Pathog 2021; 13:54. [PMID: 34517895 PMCID: PMC8439029 DOI: 10.1186/s13099-021-00448-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 08/04/2021] [Indexed: 02/08/2023] Open
Abstract
Background Innovative research highlighted the probable connection between autism spectrum disorder (ASD) and gut microbiota as many autistic individuals have gastrointestinal problems as co-morbidities. This review emphasizes the role of altered gut microbiota observed frequently in autistic patients, and the mechanisms through which such alterations may trigger leaky gut. Main body Different bacterial metabolite levels in the blood and urine of autistic children, such as short-chain fatty acids, lipopolysaccharides, beta-cresol, and bacterial toxins, were reviewed. Moreover, the importance of selected proteins, among which are calprotectin, zonulin, and lysozyme, were discussed as biomarkers for the early detection of leaky gut as an etiological mechanism of ASD through the less integrative gut–blood–brain barriers. Disrupted gut–blood–brain barriers can explain the leakage of bacterial metabolites in these patients. Conclusion Although the cause-to-effect relationship between ASD and altered gut microbiota is not yet well understood, this review shows that with the consumption of specific diets, definite probiotics may represent a noninvasive tool to reestablish healthy gut microbiota and stimulate gut health. The diagnostic and therapeutic value of intestinal proteins and bacterial-derived compounds as new possible biomarkers, as well as potential therapeutic targets, are discussed. Supplementary Information The online version contains supplementary material available at 10.1186/s13099-021-00448-y.
Collapse
Affiliation(s)
- Laila Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.,Autism Research and Treatment Center, Riyadh, Saudi Arabia
| | - Naima Zayed
- Therapuetic Chemistry Department, National Research Centre, Dokki, Cairo, Egypt
| | - Ramesa Shafi Bhat
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Nadine M S Moubayed
- Botany and Microbiology Department, College of Science, Female Campus, King Saud University, Riyadh, Saudi Arabia
| | - May N Al-Muammar
- Department of Community Health, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, P.O box 22452, Zip code 11495, Riyadh, Saudi Arabia.
| |
Collapse
|
18
|
Lerner A, Freire de Carvalho J, Kotrova A, Shoenfeld Y. Gluten-free diet can ameliorate the symptoms of non-celiac autoimmune diseases. Nutr Rev 2021; 80:525-543. [PMID: 34338776 DOI: 10.1093/nutrit/nuab039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/05/2021] [Accepted: 05/12/2021] [Indexed: 11/12/2022] Open
Abstract
CONTEXT A gluten-free diet (GFD) is the recommended treatment for gluten-dependent disease. In addition, gluten withdrawal is popular and occasionally is suggested as a treatment for other autoimmune diseases (ADs). OBJECTIVE The current systematic review summarizes those entities and discusses the logic behind using a GFD in classical non-gluten-dependentADs. DATA SOURCES A search for medical articles in PubMed/MEDLINE, Web of Sciences, LILACS, and Scielo published between 1960 and 2020 was conducted, using the key words for various ADs and GFDs. DATA EXXTRACTION Eight-three articles were included in the systematic review (using PRISMA guidelines). DATA ANALYSIS Reduction in symptoms of ADs after observance of a GFD was observed in 911 out of 1408 patients (64.7%) and in 66 out of the 83 selected studies (79.5%). The age of the patients ranged from 9 months to 69 years. The duration of the GFD varied from 1 month to 9 years. A GFD can suppress several harmful intraluminal intestinal events. Potential mechanisms and pathways for the action of GFD in the gut - remote organs' axis have been suggested. CONCLUSION A GFD might represent a novel nutritional therapeutic strategy for classical non-gluten-dependent autoimmune conditions.
Collapse
Affiliation(s)
- Aaron Lerner
- A. Lerner and Y. Shoenfeld are with the The Zabludowicz Research Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel Hashomer, Israel. J. Freire de Carvalho is with the Department of Rheumatology, Institute for Health Sciences of the Federal University of Bahia, Salvador, Bahia, Brazil. A. Kotrova and Y. Shoenfeld are with the Department of Autoimmune research, Saint Petersburg State University, St. Petersburg, Russia. Y. Shoenfeld is with the Department of Administration, Ariel University, Israel. Y. Shoenfeld is with the Department of Autoimmune research, I.M Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Jozélio Freire de Carvalho
- A. Lerner and Y. Shoenfeld are with the The Zabludowicz Research Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel Hashomer, Israel. J. Freire de Carvalho is with the Department of Rheumatology, Institute for Health Sciences of the Federal University of Bahia, Salvador, Bahia, Brazil. A. Kotrova and Y. Shoenfeld are with the Department of Autoimmune research, Saint Petersburg State University, St. Petersburg, Russia. Y. Shoenfeld is with the Department of Administration, Ariel University, Israel. Y. Shoenfeld is with the Department of Autoimmune research, I.M Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Anna Kotrova
- A. Lerner and Y. Shoenfeld are with the The Zabludowicz Research Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel Hashomer, Israel. J. Freire de Carvalho is with the Department of Rheumatology, Institute for Health Sciences of the Federal University of Bahia, Salvador, Bahia, Brazil. A. Kotrova and Y. Shoenfeld are with the Department of Autoimmune research, Saint Petersburg State University, St. Petersburg, Russia. Y. Shoenfeld is with the Department of Administration, Ariel University, Israel. Y. Shoenfeld is with the Department of Autoimmune research, I.M Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Yehuda Shoenfeld
- A. Lerner and Y. Shoenfeld are with the The Zabludowicz Research Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel Hashomer, Israel. J. Freire de Carvalho is with the Department of Rheumatology, Institute for Health Sciences of the Federal University of Bahia, Salvador, Bahia, Brazil. A. Kotrova and Y. Shoenfeld are with the Department of Autoimmune research, Saint Petersburg State University, St. Petersburg, Russia. Y. Shoenfeld is with the Department of Administration, Ariel University, Israel. Y. Shoenfeld is with the Department of Autoimmune research, I.M Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
19
|
Lerner A, Sobolevskaia P, Churilov L, Shoenfeld Y. Alpha-enolase involvement in intestinal and extraintestinal manifestations of celiac disease. J Transl Autoimmun 2021; 4:100109. [PMID: 34189450 PMCID: PMC8219987 DOI: 10.1016/j.jtauto.2021.100109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 01/13/2023] Open
Abstract
Celiac disease is a life-long intestinal autoimmune disease, characterized by the gluten intolerance and chronic enteric inflammation. Traditionally presented by intestinal manifestations, however, a shift toward extra intestinal presentation is taking place. One of the affected organs is the nervous systems presented by neuropsychiatric manifestations, hence the mechanism and pathways are not clear. The presence of neuronal and alpha-enolases and their corresponding antibodies were noticed in the mucosa and serum of celiac disease patients, as well as in other various autoimmune diseases with psycho-neurological manifestations. The aims of the present review are to screen the literature on different isoforms of enolase, mainly alpha enolase, and their specific antibodies and to suggest their potential pathophysiological mechanisms relaying the enolases to intestinal or extraintestinal celiac disease manifestations. The shared aspects between the enolases and celiac disease and the cross-talks between alpha-enolase and tissue transglutaminase suggest new potential pathophysiological mechanisms that might drive celiac disease evolvement.
Collapse
Affiliation(s)
- Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| | | | | | - Yehuda Shoenfeld
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Visiting Professor), Moscow, Russia
- Ariel University, Ariel, Israel
| |
Collapse
|
20
|
Murdaca G, Greco M, Borro M, Gangemi S. Hygiene hypothesis and autoimmune diseases: A narrative review of clinical evidences and mechanisms. Autoimmun Rev 2021; 20:102845. [PMID: 33971339 DOI: 10.1016/j.autrev.2021.102845] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/20/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022]
Abstract
Since the start of the "modern era", characterized by the increase in urbanization, a progressive attention to hygiene and autoimmune conditions has considerably grown. Although these diseases are often multifactorial, it was demonstrated that environment factors such as pollution, diet and lifestyles may play a crucial role together with genetic signature. Our research, based on the newest and most significant literature of this topic, highlights that the progressive depletion of microbes and parasites due to increased socioeconomic improvement, may lead to a derangement of immunoregulatory mechanisms. Moreover, special attention was given to the complex interplay between microbial agents, as gut microbiome, diet and vitamin D supplementation with the aim of identifying promising future therapeutic options. In conclusion, autoimmunity cannot be limited to hygiene-hypothesis, but from the point of view of precision medicine, this theory represents a fundamental element together with the study of genomics, the microbiome and proteomics, in order to understand the complex functioning of the immune system.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Monica Greco
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy
| | - Matteo Borro
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
21
|
Vojdani A, Lerner A, Vojdani E. Cross-Reactivity and Sequence Homology Between Alpha-Synuclein and Food Products: A Step Further for Parkinson's Disease Synucleinopathy. Cells 2021; 10:1111. [PMID: 34063062 PMCID: PMC8147930 DOI: 10.3390/cells10051111] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/24/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Parkinson's disease is characterized by non-motor/motor dysfunction midbrain neuronal death and α-synuclein deposits. The accepted hypothesis is that unknown environmental factors induce α-synuclein accumulation in the brain via the enteric nervous system. MATERIAL AND METHODS Monoclonal antibodies made against recombinant α-synuclein protein or α-synuclein epitope 118-123 were applied to the antigens of 180 frequently consumed food products. The specificity of those antibody-antigen reactions was confirmed by serial dilution and inhibition studies. The Basic Local Alignment Search Tool sequence matching program was used for sequence homologies. RESULTS While the antibody made against recombinant α-synuclein reacted significantly with 86/180 specific food antigens, the antibody made against α-synuclein epitope 118-123 reacted with only 32/180 tested food antigens. The food proteins with the greatest number of peptides that matched with α-synuclein were yeast, soybean, latex hevein, wheat germ agglutinin, potato, peanut, bean agglutinin, pea lectin, shrimp, bromelain, and lentil lectin. Conclusions: The cross-reactivity and sequence homology between α-synuclein and frequently consumed foods, reinforces the autoimmune aspect of Parkinson's disease. It is hypothesized that luminal food peptides that share cross-reactive epitopes with human α-synuclein and have molecular similarity with brain antigens are involved in the synucleinopathy. The findings deserve further confirmation by extensive research.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Laboratory, Inc., Los Angeles, CA 90035, USA;
- Cyrex Laboratories, Phoenix, AZ 85034, USA
- Department of Preventive Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Aaron Lerner
- Chaim Sheba Medical Center, Zabludowicz Center for Autoimmune Diseases, Tel-Hashomer 52621, Israel
| | - Elroy Vojdani
- Regenera Medical,11620 Wilshire Blvd., Ste. 470, Los Angeles, CA 90025, USA;
| |
Collapse
|
22
|
Lv L, Jiang H, Chen Y, Gu S, Xia J, Zhang H, Lu Y, Yan R, Li L. The faecal metabolome in COVID-19 patients is altered and associated with clinical features and gut microbes. Anal Chim Acta 2021; 1152:338267. [PMID: 33648648 PMCID: PMC7847702 DOI: 10.1016/j.aca.2021.338267] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
Although SARS-CoV-2 can invade the intestine, though its effect on digestion and absorption is not fully understood. In the present study, 56 COVID-19 patients and 47 age- and sex-matched healthy subjects were divided into a discovery cohort and a validation cohort. Blood, faeces and clinical information were collected from the patients in the hospital and at discharge. The faecal metabolome was analysed using gas chromatography-mass spectrometry, and Spearman's correlation analyses of clinical features, the serum metabolome, and the faecal micro- and mycobiota were conducted. The results showed that, the faeces of COVID-19 patients were enriched with important nutrients that should be metabolized or absorbed, such as sucrose and 2-palmitoyl-glycerol; diet-related components that cannot be synthesized by humans, such as 1,5-anhydroglucitol and D-pinitol; and harmful metabolites, such as oxalate, were also detected. In contrast, purine metabolites such as deoxyinosine and hypoxanthine, low-water-soluble long-chain fatty alcohols/acids such as behenic acid, compounds rarely occurring in nature such as D-allose and D-arabinose, and microbe-related compounds such as 2,4-di-tert-butylphenol were depleted in the faeces of COVID-19 patients. Moreover, these metabolites significantly correlated with altered serum metabolites such as oxalate and gut microbesincluding Ruminococcaceae, Actinomyces, Sphingomonas and Aspergillus. Although levels of several faecal metabolites, such as sucrose, 1,5-anhydroglucitol and D-pinitol, of discharged patients were not different from those of healthy controls (HCs), those of oxalate and 2-palmitoyl-glycerol did differ. Therefore, alterations in the faecal metabolome of COVID-19 patients may reflect malnutrition and intestinal inflammation and warrant greater attention. The results of present study provide new insights into the pathogenesis and treatment of COVID-19.
Collapse
Affiliation(s)
- Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Huiyong Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Silan Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Hua Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Yingfeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Ren Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China.
| |
Collapse
|
23
|
Lerner A, Benzvi C. "Let Food Be Thy Medicine": Gluten and Potential Role in Neurodegeneration. Cells 2021; 10:756. [PMID: 33808124 PMCID: PMC8065505 DOI: 10.3390/cells10040756] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Wheat is a most favored staple food worldwide and its major protein is gluten. It is involved in several gluten dependent diseases and lately was suggested to play a role in non-celiac autoimmune diseases. Its involvement in neurodegenerative conditions was recently suggested but no cause-and-effect relationship were established. The present narrative review expands on various aspects of the gluten-gut-brain axes events, mechanisms and pathways that connect wheat and gluten consumption to neurodegenerative disease. Gluten induced dysbiosis, increased intestinal permeabillity, enteric and systemic side effects, cross-reactive antibodies, and the sequence of homologies between brain antigens and gluten are highlighted. This combination may suggest molecular mimicry, alluding to some autoimmune aspects between gluten and neurodegenerative disease. The proverb of Hippocrates coined in 400 BC, "let food be thy medicine," is critically discussed in the frame of gluten and potential neurodegeneration evolvement.
Collapse
Affiliation(s)
- Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer 5262000, Israel;
| | | |
Collapse
|
24
|
Duncanson K, Burns G, Pryor J, Keely S, Talley NJ. Mechanisms of Food-Induced Symptom Induction and Dietary Management in Functional Dyspepsia. Nutrients 2021; 13:1109. [PMID: 33800668 PMCID: PMC8066021 DOI: 10.3390/nu13041109] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Functional dyspepsia (FD) is a common disorder of gut-brain interaction, characterised by upper gastrointestinal symptom profiles that differentiate FD from the irritable bowel syndrome (IBS), although the two conditions often co-exist. Despite food and eating being implicated in FD symptom induction, evidence-based guidance for dietetic management of FD is limited. The aim of this narrative review is to collate the possible mechanisms for eating-induced and food-related symptoms of FD for stratification of dietetic management. Specific carbohydrates, proteins and fats, or foods high in these macronutrients have all been reported as influencing FD symptom induction, with removal of 'trigger' foods or nutrients shown to alleviate symptoms. Food additives and natural food chemicals have also been implicated, but there is a lack of convincing evidence. Emerging evidence suggests the gastrointestinal microbiota is the primary interface between food and symptom induction in FD, and is therefore a research direction that warrants substantial attention. Objective markers of FD, along with more sensitive and specific dietary assessment tools will contribute to progressing towards evidence-based dietetic management of FD.
Collapse
Affiliation(s)
- Kerith Duncanson
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (G.B.); (J.P.); (S.K.)
- Centre for Research Excellence, Digestive Health, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Grace Burns
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (G.B.); (J.P.); (S.K.)
- Centre for Research Excellence, Digestive Health, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Jennifer Pryor
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (G.B.); (J.P.); (S.K.)
- Centre for Research Excellence, Digestive Health, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Simon Keely
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (G.B.); (J.P.); (S.K.)
- Centre for Research Excellence, Digestive Health, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Nicholas J. Talley
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (G.B.); (J.P.); (S.K.)
- Centre for Research Excellence, Digestive Health, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- Department of Gastroenterology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
25
|
Ren R, Lu D, Liu T. Development of a sandwich-type rat small intestine tissue sensor for detecting resveratrol and its receptors. Biomed Microdevices 2021; 23:13. [PMID: 33666776 DOI: 10.1007/s10544-021-00554-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
Resveratrol has a variety of biological functions, however, a limited number of studies have assessed its interaction with cell surface receptors. In this study, a sandwich-type rat small intestine tissue sensor (RSIT-sensor) was fabricated to detect the response current from receptor stimulation by different resveratrol concentrations via electrochemical workstation. The results showed that with detection limit of 1 × 10-13 mol/L, the maximum rate of change of the response current was found at the concentration of 8.5 × 10-12 mol/L, indicating that the resveratrol-related receptor was saturated. With comparing the response values of prepared biosensor and bare electrode with resveratrol, it can be concluded that the response value of small intestinal cells to resveratrol has obviously been amplified by the intracellular signal transmission system, and its magnification was about 100 times. In the current research, for the first time, kinetics of the interaction between resveratrol and its receptors and the transmission of signals to the body could be quantitatively measured by a biosensor. Our findings may provide new ideas for resveratrol-related receptor analysis, separation and purification, signal transmission, and evaluation of biological function.
Collapse
Affiliation(s)
- Ruijuan Ren
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, 300314, China
| | - Dingqiang Lu
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, 300314, China. .,Tianjin Key Laboratory of Food Biotechnology, Tianjin, 300314, China.
| | - Tingting Liu
- College of Biotechnology & Food Science, Tianjin University of Commerce, Tianjin, 300314, China
| |
Collapse
|
26
|
Cassotta M, Forbes-Hernandez TY, Cianciosi D, Elexpuru Zabaleta M, Sumalla Cano S, Dominguez I, Bullon B, Regolo L, Alvarez-Suarez JM, Giampieri F, Battino M. Nutrition and Rheumatoid Arthritis in the 'Omics' Era. Nutrients 2021; 13:763. [PMID: 33652915 PMCID: PMC7996781 DOI: 10.3390/nu13030763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/16/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Modern high-throughput 'omics' science tools (including genomics, transcriptomics, proteomics, metabolomics and microbiomics) are currently being applied to nutritional sciences to unravel the fundamental processes of health effects ascribed to particular nutrients in humans and to contribute to more precise nutritional advice. Diet and food components are key environmental factors that interact with the genome, transcriptome, proteome, metabolome and the microbiota, and this life-long interplay defines health and diseases state of the individual. Rheumatoid arthritis (RA) is a chronic autoimmune disease featured by a systemic immune-inflammatory response, in genetically susceptible individuals exposed to environmental triggers, including diet. In recent years increasing evidences suggested that nutritional factors and gut microbiome have a central role in RA risk and progression. The aim of this review is to summarize the main and most recent applications of 'omics' technologies in human nutrition and in RA research, examining the possible influences of some nutrients and nutritional patterns on RA pathogenesis, following a nutrigenomics approach. The opportunities and challenges of novel 'omics technologies' in the exploration of new avenues in RA and nutritional research to prevent and manage RA will be also discussed.
Collapse
Affiliation(s)
- Manuela Cassotta
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (M.C.); (M.E.Z.); (S.S.C.); (I.D.)
| | - Tamara Y. Forbes-Hernandez
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
| | - Danila Cianciosi
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy; (D.C.); (L.R.)
| | - Maria Elexpuru Zabaleta
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (M.C.); (M.E.Z.); (S.S.C.); (I.D.)
| | - Sandra Sumalla Cano
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (M.C.); (M.E.Z.); (S.S.C.); (I.D.)
| | - Irma Dominguez
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain; (M.C.); (M.E.Z.); (S.S.C.); (I.D.)
| | - Beatriz Bullon
- Department of Periodontology, Dental School, University of Sevilla, 41004 Sevilla, Spain;
| | - Lucia Regolo
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy; (D.C.); (L.R.)
| | - Josè Miguel Alvarez-Suarez
- AgroScience & Food Research Group, Universidad de Las Américas, Quito 170125, Ecuador;
- King Fahd Medical Research Center, King Abdulaziz University, Jedda 21589, Saudi Arabia
| | - Francesca Giampieri
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy; (D.C.); (L.R.)
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Maurizio Battino
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy; (D.C.); (L.R.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
27
|
Vojdani A, Turnpaugh CC. Antibodies against Group A Streptococcus, dopamine receptors, and ganglioside GM1 cross-react with a variety of food antigens, potentially interfering with biomarkers for PANS and PANDAS. Biomark Neuropsychiatry 2020. [DOI: 10.1016/j.bionps.2020.100023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|