1
|
Abdelhamed HG, Hassan AA, Sakraan AA, Al-Deeb RT, Mousa DM, Aboul Ezz HS, Noor NA, Khadrawy YA, Radwan NM. Brain interleukins and Alzheimer's disease. Metab Brain Dis 2025; 40:116. [PMID: 39891777 PMCID: PMC11787210 DOI: 10.1007/s11011-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
The central nervous system (CNS) is immune-privileged by several immuno-modulators as interleukins (ILs). ILs are cytokines secreted by immune cells for cell-cell signaling communications and affect the functions of the CNS. ILs were reported to orchestrate different molecular and cellular mechanisms of both physiological and pathological events, through overproduction or over-expression of their receptors. They interact with numerous receptors mediating pro-inflammatory and/or anti-inflammatory actions. Interleukins have been implicated to participate in neurodegenerative diseases. They play a critical role in Alzheimer's disease (AD) pathology which is characterized by the over-production of pro-inflammatory ILs. These may aggravate neurodegeneration, in addition to their contribution to detrimental mechanisms as oxidative stress, and excitotoxicity. However, recent research on the relation between ILs and AD revealed major discrepancies. Most of the major ILs were shown to play both pro- and anti-inflammatory roles in different experimental settings and models. The interactions between different ILs through shared pathways also add to the difficulty of drawing solid conclusions. In addition, targeting the different ILs has not yielded consistent results. The repeated failures of therapeutic drugs in treating AD necessitate the search for novel agents targeting multiple mechanisms of the disease pathology. In this context, the understanding of interleukins and their roles throughout the disease progression and interaction with other systems in the brain may provide promising therapeutic targets for the prevention or treatment of AD.
Collapse
Affiliation(s)
- Heba G Abdelhamed
- Department of Zoology and Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Arwa A Hassan
- Faculty of Pharmacy & Pharmaceutical Industries, Sinai University, Sinai, Egypt
| | - Alaa A Sakraan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Dalia M Mousa
- Department of Biotechnology, Faculty of Science, Cairo University, Giza, Egypt
| | - Heba S Aboul Ezz
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Neveen A Noor
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Nasr M Radwan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
2
|
Bertolini M, Clark D. Periodontal disease as a model to study chronic inflammation in aging. GeroScience 2024; 46:3695-3709. [PMID: 37285008 PMCID: PMC11226587 DOI: 10.1007/s11357-023-00835-0] [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: 01/27/2023] [Accepted: 05/20/2023] [Indexed: 06/08/2023] Open
Abstract
Periodontal disease is a chronic inflammatory condition that results in the destruction of the teeth supporting tissues, eventually leading to the loss of teeth and reduced quality of life. In severe cases, periodontal disease can limit proper nutritional intake, cause acute pain and infection, and cause a withdrawal from social situations due to esthetic and phonetic concerns. Similar to other chronic inflammatory conditions, periodontal disease increases in prevalence with age. Research into what drives periodontal disease pathogenesis in older adults is contributing to our general understanding of age-related chronic inflammation. This review will present periodontal disease as an age-related chronic inflammatory disease and as an effective geroscience model to study mechanisms of age-related inflammatory dysregulation. The current understanding of the cellular and molecular mechanisms that drive inflammatory dysregulation as a function of age will be discussed with a focus on the major pathogenic immune cells in periodontal disease, which include neutrophils, macrophages, and T cells. Research in the aging biology field has shown that the age-related changes in these immune cells result in the cells becoming less effective in the clearance of microbial pathogens, expansion of pathogenic subpopulations, or an increase in pro-inflammatory cytokine secretions. Such changes can be pathogenic and contribute to inflammatory dysregulation that is associated with a myriad of age-related disease including periodontal disease. An improved understanding is needed to develop better interventions that target the molecules or pathways that are perturbed with age in order to improve treatment of chronic inflammatory conditions, including periodontal disease, in older adult populations.
Collapse
Affiliation(s)
- Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Daniel Clark
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Singh J, Jackson KL, Tang FS, Fu T, Nowell C, Salimova E, Kiriazis H, Ritchie RH, Head GA, Woodman OL, Qin CX. The pro-resolving mediator, annexin A1 regulates blood pressure, and age-associated changes in cardiovascular function and remodeling. FASEB J 2024; 38:e23457. [PMID: 38318648 DOI: 10.1096/fj.202301802r] [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: 09/05/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024]
Abstract
Aging is associated with chronic, low-level inflammation which may contribute to cardiovascular pathologies such as hypertension and atherosclerosis. This chronic inflammation may be opposed by endogenous mechanisms to limit inflammation, for example, by the actions of annexin A1 (ANXA1), an endogenous glucocorticoid-regulated protein that has anti-inflammatory and pro-resolving activity. We hypothesized the pro-resolving mediator ANXA1 protects against age-induced changes in blood pressure (BP), cardiovascular structure and function, and cardiac senescence. BP was measured monthly in conscious mature (4-month) and middle-aged (12-month) ANXA1-deficient (ANXA1-/- ) and wild-type C57BL/6 mice. Body composition was measured using EchoMRI, and both cardiac and vascular function using ultrasound imaging. Cardiac hypertrophy, fibrosis and senescence, vascular fibrosis, elastin, and calcification were assessed histologically. Gene expression relevant to structural remodeling, inflammation, and cardiomyocyte senescence were also quantified. In C57BL/6 mice, progression from 4 to 12 months of age did not affect the majority of cardiovascular parameters measured, with the exception of mild cardiac hypertrophy, vascular calcium, and collagen deposition. Interestingly, ANXA1-/- mice exhibited higher BP, regardless of age. Additionally, age progression had a marked impact in ANXA1-/- mice, with markedly augmented vascular remodeling, impaired vascular distensibility, and body composition. Consistent with vascular dysfunction, cardiac dysfunction, and hypertrophy were also evident, together with markers of senescence and inflammation. These findings suggest that endogenous ANXA1 plays a critical role in regulating BP, cardiovascular function, and remodeling and delays cardiac senescence. Our findings support the development of novel ANXA1-based therapies to prevent age-related cardiovascular pathologies.
Collapse
Affiliation(s)
- Jaideep Singh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Kristy L Jackson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Feng Shii Tang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ting Fu
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ekaterina Salimova
- Monash Biomedical Imaging, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Geoffrey A Head
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Owen L Woodman
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cheng Xue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Pharmacology, School of Pharmaceutical Sciences, Qilu College of Medicine, Shandong University, Jinan, China
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
4
|
Leal VNC, Paulino LM, Cambui RAG, Zupelli TG, Yamada SM, Oliveira LAT, Dutra VDF, Bub CB, Sakashita AM, Yokoyama APH, Kutner JM, Vieira CA, Santiago WMDS, Andrade MMS, Teixeira FME, Alberca RW, Gozzi-Silva SC, Yendo TM, Netto LC, Duarte AJS, Sato MN, Venturini J, Pontillo A. A common variant close to the "tripwire" linker region of NLRP1 contributes to severe COVID-19. Inflamm Res 2023; 72:1933-1940. [PMID: 36416944 PMCID: PMC9684769 DOI: 10.1007/s00011-022-01670-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/26/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE AND DESIGN The heterogeneity of response to SARS-CoV-2 infection is directly linked to the individual genetic background. Genetic variants of inflammasome-related genes have been pointed as risk factors for several inflammatory sterile and infectious disease. In the group of inflammasome receptors, NLRP1 stands out as a good novel candidate as severity factor for COVID-19 disease. METHODS To address this question, we performed an association study of NLRP1, DPP9, CARD8, IL1B, and IL18 single nucleotide variants (SNVs) in a cohort of 945 COVID-19 patients. RESULTS The NLRP1 p.Leu155His in the linker region, target of viral protease, was significantly associated to COVID-19 severity, which could contribute to the excessive cytokine release reported in severe cases. CONCLUSION Inflammasome genetic background contributes to individual response to SARS-CoV-2.
Collapse
Affiliation(s)
- Vinicius N C Leal
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, SP, Brasil
| | - Leandro M Paulino
- Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul, Campo grande, Mato Grosso do Sul, Brasil
| | - Raylane A G Cambui
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, SP, Brasil
| | - Thiago G Zupelli
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, SP, Brasil
| | - Suemy M Yamada
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, SP, Brasil
| | - Leonardo A T Oliveira
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, SP, Brasil
| | - Valéria de F Dutra
- Departamento de Hemoterapia, Hospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - Carolina B Bub
- Departamento de Hemoterapia, Hospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - Araci M Sakashita
- Departamento de Hemoterapia, Hospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - Ana Paula H Yokoyama
- Departamento de Hemoterapia, Hospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - José M Kutner
- Departamento de Hemoterapia, Hospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - Camila A Vieira
- Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul, Campo grande, Mato Grosso do Sul, Brasil
| | - Wellyngton M de S Santiago
- Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul, Campo grande, Mato Grosso do Sul, Brasil
| | - Milena M S Andrade
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56, Departamento de Dermatologia, Hospital das Clínicas E Faculdade de Medicina/HCFMUSP, São Paulo, Brasil
| | - Franciane M E Teixeira
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56, Departamento de Dermatologia, Hospital das Clínicas E Faculdade de Medicina/HCFMUSP, São Paulo, Brasil
| | - Ricardo W Alberca
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56, Departamento de Dermatologia, Hospital das Clínicas E Faculdade de Medicina/HCFMUSP, São Paulo, Brasil
| | - Sarah C Gozzi-Silva
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56, Departamento de Dermatologia, Hospital das Clínicas E Faculdade de Medicina/HCFMUSP, São Paulo, Brasil
| | - Tatiana M Yendo
- Departamento de Dermatologia, Faculdade de Medicina, Instituto de Medicina Tropical, Universidade de São Paulo/FMUSP, São Paulo, Brasil
| | - Lucas C Netto
- Unidade Terapia Intensiva, Hospital das Clínicas/FMUSP, São Paulo, Brasil
| | - Alberto J S Duarte
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56, Departamento de Dermatologia, Hospital das Clínicas E Faculdade de Medicina/HCFMUSP, São Paulo, Brasil
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo/FMUSP, São Paulo, Brasil
| | - Maria N Sato
- Laboratório de Investigação Médica em Dermatologia e Imunodeficiências (LIM-56, Departamento de Dermatologia, Hospital das Clínicas E Faculdade de Medicina/HCFMUSP, São Paulo, Brasil
| | - James Venturini
- Faculdade de Medicina, Universidade Federal de Mato Grosso do Sul, Campo grande, Mato Grosso do Sul, Brasil
| | - Alessandra Pontillo
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas/ICB, Universidade de São Paulo/USP, São Paulo, SP, Brasil.
| |
Collapse
|
5
|
Vicente MC, Paneghini JL, Stabile AM, Amorim M, Anibal Silva CE, Patrone LGA, Cunha TM, Bícego KC, Almeida MC, Carrettiero DC, Gargaglioni LH. Inhibition of Pro-Inflammatory Microglia with Minocycline Improves Cognitive and Sleep-Wake Dysfunction Under Respiratory Stress in a Sporadic Model for Alzheimer's Disease. J Alzheimers Dis 2023; 95:317-337. [PMID: 37522205 DOI: 10.3233/jad-230151] [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] [Indexed: 08/01/2023]
Abstract
BACKGROUND Neuroinflammation in Alzheimer's disease (AD) can occur due to excessive activation of microglia in response to the accumulation of amyloid-β peptide (Aβ). Previously, we demonstrated an increased expression of this peptide in the locus coeruleus (LC) in a sporadic model for AD (streptozotocin, STZ; 2 mg/kg, ICV). We hypothesized that the STZ-AD model exhibits neuroinflammation, and treatment with an inhibitor of microglia (minocycline) can reverse the cognitive, respiratory, sleep, and molecular disorders of this model. OBJECTIVE To evaluate the effect of minocycline treatment in STZ model disorders. METHODS We treated control and STZ-treated rats for five days with minocycline (30 mg/kg, IP) and evaluated cognitive performance, chemoreflex response to hypercapnia and hypoxia, and total sleep time. Additionally, quantification of Aβ, microglia analyses, and relative expression of cytokines in the LC were performed. RESULTS Minocycline treatment improved learning and memory, which was concomitant with a decrease in microglial cell density and re-establishment of morphological changes induced by STZ in the LC region. Minocycline did not reverse the STZ-induced increase in CO2 sensitivity during wakefulness. However, it restored the daytime sleep-wake cycle in STZ-treated animals to the same levels as those observed in control animals. In the LC, levels of A and expression of Il10, Il1b, and Mcp1 mRNA remained unaffected by minocycline, but we found a strong trend of minocycline effect on Tnf- α. CONCLUSION Our findings suggest that minocycline effectively reduces microglial recruitment and the inflammatory morphological profile in the LC, while it recovers cognitive performance and restores the sleep-wake pattern impaired by STZ.
Collapse
Affiliation(s)
- Mariane C Vicente
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Julia L Paneghini
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Angelita M Stabile
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mateus Amorim
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Conceição E Anibal Silva
- Department of Pharmachology, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Thiago M Cunha
- Department of Pharmachology, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Maria C Almeida
- Center for Natural and Human Sciences, Federal University of ABC, São Bernardo do Campo, Brazil
| | - Daniel C Carrettiero
- Center for Natural and Human Sciences, Federal University of ABC, São Bernardo do Campo, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV, Jaboticabal, SP, Brazil
| |
Collapse
|
6
|
Behzadi P, Sameer AS, Nissar S, Banday MZ, Gajdács M, García-Perdomo HA, Akhtar K, Pinheiro M, Magnusson P, Sarshar M, Ambrosi C. The Interleukin-1 (IL-1) Superfamily Cytokines and Their Single Nucleotide Polymorphisms (SNPs). J Immunol Res 2022; 2022:2054431. [PMID: 35378905 PMCID: PMC8976653 DOI: 10.1155/2022/2054431] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/24/2022] [Accepted: 03/08/2022] [Indexed: 12/19/2022] Open
Abstract
Interleukins (ILs)-which are important members of cytokines-consist of a vast group of molecules, including a wide range of immune mediators that contribute to the immunological responses of many cells and tissues. ILs are immune-glycoproteins, which directly contribute to the growth, activation, adhesion, differentiation, migration, proliferation, and maturation of immune cells; and subsequently, they are involved in the pro and anti-inflammatory responses of the body, by their interaction with a wide range of receptors. Due to the importance of immune system in different organisms, the genes belonging to immune elements, such as ILs, have been studied vigorously. The results of recent investigations showed that the genes pertaining to the immune system undergo progressive evolution with a constant rate. The occurrence of any mutation or polymorphism in IL genes may result in substantial changes in their biology and function and may be associated with a wide range of diseases and disorders. Among these abnormalities, single nucleotide polymorphisms (SNPs) can represent as important disruptive factors. The present review aims at concisely summarizing the current knowledge available on the occurrence, properties, role, and biological consequences of SNPs within the IL-1 family members.
Collapse
Affiliation(s)
- Payam Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran 37541-374, Iran
| | - Aga Syed Sameer
- Molecular Disease & Diagnosis Division, Infinity Biochemistry Pvt. Ltd, Sajjad Abad, Chattabal, Srinagar, Kashmir, India
- Department of Biochemistry, Government Medical College, Karan Nagar, Srinagar, Kashmir, India
| | - Saniya Nissar
- Molecular Disease & Diagnosis Division, Infinity Biochemistry Pvt. Ltd, Sajjad Abad, Chattabal, Srinagar, Kashmir, India
- Department of Biochemistry, Government Medical College, Karan Nagar, Srinagar, Kashmir, India
| | - Mujeeb Zafar Banday
- Molecular Disease & Diagnosis Division, Infinity Biochemistry Pvt. Ltd, Sajjad Abad, Chattabal, Srinagar, Kashmir, India
- Department of Biochemistry, Government Medical College, Karan Nagar, Srinagar, Kashmir, India
| | - Márió Gajdács
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6720 Szeged, Hungary
| | - Herney Andrés García-Perdomo
- Division of Urology, Department of Surgery, School of Medicine, UROGIV Research Group, Universidad del Valle, Cali, Colombia
| | - Kulsum Akhtar
- Department of Clinical Biochemistry, Sher I Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, India
| | - Marina Pinheiro
- Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- CHUP, Centro Hospitalar Universitário do Porto, Largo do Prof. Abel Salazar, 4099-001 Porto, Portugal
| | - Peter Magnusson
- School of Medical Sciences, Örebro University, SE, 701 82 Örebro, Sweden
- Cardiology Research Unit, Department of Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Cecilia Ambrosi
- IRCCS San Raffaele Roma, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| |
Collapse
|
7
|
Uddin MS, Kabir MT, Jalouli M, Rahman MA, Jeandet P, Behl T, Alexiou A, Albadrani GM, Abdel-Daim MM, Perveen A, Ashraf GM. Neuroinflammatory Signaling in the Pathogenesis of Alzheimer's Disease. Curr Neuropharmacol 2021; 20:126-146. [PMID: 34525932 PMCID: PMC9199559 DOI: 10.2174/1570159x19666210826130210] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 04/16/2021] [Accepted: 05/10/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease characterized by the formation of intracellular neurofibrillary tangles (NFTs) and extracellular amyloid plaques. Growing evidence has suggested that AD pathogenesis is not only limited to the neuronal compartment but also strongly interacts with immunological processes in the brain. On the other hand, aggregated and misfolded proteins can bind with pattern recognition receptors located on astroglia and microglia and can, in turn, induce an innate immune response, characterized by the release of inflammatory mediators, ultimately playing a role in both the severity and the progression of the disease. It has been reported by genome-wide analysis that several genes which elevate the risk for sporadic AD encode for factors controlling the inflammatory response and glial clearance of misfolded proteins. Obesity and systemic inflammation are examples of external factors which may interfere with the immunological mechanisms of the brain and can induce disease progression. In this review, we discussed the mechanisms and essential role of inflammatory signaling pathways in AD pathogenesis. Indeed, interfering with immune processes and modulation of risk factors may lead to future therapeutic or preventive AD approaches.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka. Bangladesh
| | | | - Maroua Jalouli
- College of Science, King Saud University, P.O. Box 2455, Riyadh 11451. Saudi Arabia
| | - Md Ataur Rahman
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul. Korea
| | - Philippe Jeandet
- Research Unit "Induced Resistance and Plant Bioprotection", EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims Cedex 2. France
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, 2770 Hebersham. Australia
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474. Saudi Arabia
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522. Egypt
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur. India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah. Saudi Arabia
| |
Collapse
|
8
|
AlRuwaisan AU, Al-Anazi MR, Shafeai MI, Rudiny FH, Motaen AM, Bin Dajem SM, Alothaid H, Morsy K, Alkahtani S, Al-Qahtani AA. Associations of Single Nucleotide Polymorphisms in IL-18 Gene with Plasmodium falciparum-Associated Malaria. J Inflamm Res 2021; 14:3587-3619. [PMID: 34345179 PMCID: PMC8323861 DOI: 10.2147/jir.s314638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/23/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Interleukin-18 (IL-18) is a pro-inflammatory cytokine, reported to be involved in the initial immune responses against malaria. Genetic variations in the host are an important factor that influences the etiology of malaria at several disease levels. Polymorphisms within the IL-18 gene are associated with susceptibility and clinical outcome of several diseases. Methods We genotyped single nucleotide polymorphisms (SNPs) in IL-18 of patients infected with Plasmodium falciparum with varying extent of parasitemia and different age groups. Results SNPs rs5744292 (OR = 70.446; 95% CI = 4.318-1149.323; p<0.0001) and rs544354 (OR = 1.498; 95% CI = 1.088-2.063; p=0.013) were found to be significantly associated with parasitemia in P. falciparum-infected patients when compared with healthy control subjects. SNP rs5744292 (OR = 7.597; 95% CI=1.028-56.156; p=0.019) was associated with increased parasite density in infected patients. SNPs rs544354 (OR 0.407; 95% CI=0.204-0.812; p = 0.009) and rs360714 (OR of 0.256; 95% CI=0.119-0.554; p = 0.001) were significantly associated with parasite density in an age-dependent manner, with the risk alleles present more frequently among the younger (1-9 years) patients. Several haplotypes were found to have a significant association with parasitemia. In-vitro expression analysis using luciferase reporter assay showed that SNPs rs1946518 and rs187238 in the IL-18 gene promoter region and rs360728 and rs5744292 in the 3'-untranslated region of the IL-18 gene were associated with enhanced transcriptional activity. Conclusion Our results suggest that polymorphisms within the IL-18 gene are associated with the susceptibility to P. falciparum infection and related parasitemia among groups with different parasite density and across various age groups.
Collapse
Affiliation(s)
- Alaa U AlRuwaisan
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mashael R Al-Anazi
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | | | | | | | - Saad M Bin Dajem
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Saad Alkahtani
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Kelly LA, O'Dea MI, Zareen Z, Melo AM, McKenna E, Strickland T, McEneaney V, Donoghue V, Boylan G, Sweetman D, Butler J, Vavasseur C, Miletin J, El-Khuffash AF, O'Neill LAJ, O'Leary JJ, Molloy EJ. Altered inflammasome activation in neonatal encephalopathy persists in childhood. Clin Exp Immunol 2021; 205:89-97. [PMID: 33768526 PMCID: PMC8209598 DOI: 10.1111/cei.13598] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/08/2021] [Accepted: 03/16/2021] [Indexed: 11/26/2022] Open
Abstract
Neonatal encephalopathy (NE) is characterized by altered neurological function in term infants and inflammation plays an important pathophysiological role. Inflammatory cytokines interleukin (IL)‐1β, IL‐1ra and IL‐18 are activated by the nucleotide‐binding and oligomerization domain (NOD)‐, leucine‐rich repeat domain (LRR)‐ and NOD‐like receptor protein 3 (NLRP3) inflammasome; furthermore, we aimed to examine the role of the inflammasome multiprotein complex involved in proinflammatory responses from the newborn period to childhood in NE. Cytokine concentrations were measured by multiplex enzyme‐linked immunosorbent assay (ELISA) in neonates and children with NE in the absence or presence of lipopolysaccharide (LPS) endotoxin. We then investigated expression of the NLRP3 inflammasome genes, NLRP3, IL‐1β and ASC by polymerase chain reaction (PCR). Serum samples from 40 NE patients at days 1 and 3 of the first week of life and in 37 patients at age 4–7 years were analysed. An increase in serum IL‐1ra and IL‐18 in neonates with NE on days 1 and 3 was observed compared to neonatal controls. IL‐1ra in NE was decreased to normal levels at school age, whereas serum IL‐18 in NE was even higher at school age compared to school age controls and NE in the first week of life. Percentage of LPS response was higher in newborns compared to school‐age NE. NLRP3 and IL‐1β gene expression were up‐regulated in the presence of LPS in NE neonates and NLRP3 gene expression remained up‐regulated at school age in NE patients compared to controls. Increased inflammasome activation in the first day of life in NE persists in childhood, and may increase the window for therapeutic intervention.
Collapse
Affiliation(s)
- L A Kelly
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - M I O'Dea
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - Z Zareen
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland.,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - A M Melo
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - E McKenna
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - T Strickland
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - V McEneaney
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - V Donoghue
- Radiology, National Maternity Hospital, Dublin, Ireland
| | - G Boylan
- Department of Pediatrics and Child Health, University College Cork, Cork, Ireland.,Infant Research Centre, Cork University Hospital, Cork, Ireland
| | - D Sweetman
- National Maternity Hospital, Dublin, Ireland
| | - J Butler
- Meso-Scale Diagnostics, Manchester, UK
| | - C Vavasseur
- National Maternity Hospital, Dublin, Ireland
| | - J Miletin
- Neonatology, Coombe Women and Infants University Hospital, Dublin, Ireland
| | | | - L A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - J J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - E J Molloy
- Discipline of Paediatrics, Trinity College, University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity College Dublin and Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland.,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland.,Neonatology, Coombe Women and Infants University Hospital, Dublin, Ireland.,CHI at Crumlin, Dublin, Ireland
| |
Collapse
|
10
|
Sathyan S, Verghese J. Genetics of frailty: A longevity perspective. Transl Res 2020; 221:83-96. [PMID: 32289255 PMCID: PMC7729977 DOI: 10.1016/j.trsl.2020.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 12/31/2022]
Abstract
Frailty is a complex late life phenotype characterized by cumulative declines in multiple physiological systems that increases the risk for disability and mortality. The biological changes associated with aging are risk factors for frailty as well as for complex diseases; whereas longevity is assumed to be an outcome of protective biological mechanisms. Understanding the interplay between biological alterations associated with aging and protective mechanisms associated with longevity in the context of frailty may help guide development of interventions to increase healthspan and promote successful aging. The complexity of these phenotypes and relatively low heritability in studies are the main roadblocks in deciphering genetic mechanisms of these age associated conditions. We review genetic research related to frailty, and discuss the possible intertwined biology of frailty and longevity.
Collapse
Affiliation(s)
- Sanish Sathyan
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
| | - Joe Verghese
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
11
|
Alipourfard I, Di Renzo L, De Lorenzo A, Khamoushi A, Cioccoloni G, Spataro F, Abbasi A. Single nucleotide polymorphism A-511 G of IL-1 gene modifies anthropometric and physiological parameters of athletes. Meta Gene 2019. [DOI: 10.1016/j.mgene.2019.100598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
12
|
Radujkovic A, Kordelas L, Dai H, Schult D, Majer-Lauterbach J, Beelen D, Müller-Tidow C, Dreger P, Luft T. Interleukin-18 and outcome after allogeneic stem cell transplantation: A retrospective cohort study. EBioMedicine 2019; 49:202-212. [PMID: 31680001 PMCID: PMC6945194 DOI: 10.1016/j.ebiom.2019.10.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin-18 (IL-18) is involved in endothelial activation and dysfunction, and in the pathogenesis and severity of acute graft-versus-host disease (aGVHD). Its relevance for patient outcome after allogeneic stem cell transplantation (alloSCT) has not yet been comprehensively addressed. Methods Pre-transplant serum levels of free IL-18 were retrospectively assessed in a cohort of 589 patients (training cohort). Results were validated in 688 patients allografted in a different centre. The primary endpoint was overall survival (OS). Secondary endpoints included incidences of non-relapse mortality (NRM), relapse, and aGVHD. Findings In the training cohort, higher pre-transplant levels of free IL-18 were significantly associated with worse OS (hazard ratio [HR] per 1-log2 increase, 1.25, P = 0.008) in multivariable models. This was due to a higher hazard of NRM (HR per 1-log2 increase, 1.39, P = 0.001), rather than relapse. The associations of pre-transplant free IL-18 with higher NRM (HR per 1-log2 increase, 1.24, P = 0.02) and shorter OS (HR per 1-log2 increase, 1.22, P = 0.006) were confirmed in the validation cohort. In both cohorts, the correlations of higher pre-transplant free IL-18 serum levels with increased NRM and worse OS were mainly driven by fatal infectious complications. No associations with incidence of aGVHD were observed. Interpretation Higher pre-transplant levels of free IL-18 were associated with non-relapse and overall mortality after alloSCT. Our results may provide a rationale for prospective studies evaluating IL-18 status and inhibition of IL-18 activity in patients undergoing allografting.
Collapse
Affiliation(s)
- Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Hao Dai
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - David Schult
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Joshua Majer-Lauterbach
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Dietrich Beelen
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany.
| |
Collapse
|
13
|
Soares JL, Oliveira EM, Pontillo A. Variants in NLRP3 and NLRC4 inflammasome associate with susceptibility and severity of multiple sclerosis. Mult Scler Relat Disord 2019; 29:26-34. [PMID: 30658261 DOI: 10.1016/j.msard.2019.01.023] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a neurodegenerative disease of central nervous system (CNS) with autoimmune and inflammatory characteristics, and a still uncertain pathogenesis. Early events as well as evolution of MS are heterogeneous (three main clinical forms) and multifactorial. Genome-wide association studies indicate that MS pathogenesis shares features with both autoimmune and inflammatory diseases. Innate immunity has been recently proved to be an important factor in MS. Genetic variants in inflammasome components have been associated with both autoimmune and neurodegenerative diseases, letting us hypothesize that inflammasome, and related cytokines IL-1ß and IL-18, could represent important contributors in MS pathogenesis, and eventually explain, at least in part, the heterogeneity observed in MS patients. AIM To evaluate the contribution of inflammasome in MS, in term of (a) genetic effect on development, severity and/or prognosis, and (b) complex activation in peripheral blood as a measure of systemic inflammation. METHODS Functional genetic variants in inflammasome components were analyzed in a cohort of MS patients, by the use of allele-specific assays and qPCR. Multivariate analysis was performed based on clinical form (recurrent remittent/RR, primary progressive/PP or secondary progressive/SP), severity index (EDSS) and progression index (PI), response to IFN-ß treatment. Peripheral blood monocytes (PBM) of patients were examined for inflammasome activation and expression profile. RESULTS AND DISCUSSION Variants associated with low serum levels of IL-18 were significantly less frequent in MS patients than in controls, suggesting a protective role of diminished IL-18-mediate inflammation in MS development. On the other hands, gain-of-function variants in NLRP3 (Q705K) and IL1B (-511 C >T) associated with severity and progression of MS, suggesting that a constitutive activation of NLRP3 inflammasome could represent a risk factor for MS clinical presentation. Accordingly, -511C >T SNP resulted more frequent in progressive forms than in RR MS, reinforcing the idea that increased inflammasome activation characterized bad prognosis of MS. Altogether these findings corroborate previous data about the harmful role of NLRP3 inflammasome in experimental autoimmune encephalitis (EAE). Moreover, we reported for the first time the beneficial effect of NLRC4 rs479333 G >C variant in MS progression and in the response to IFN-ß treatment. This intronic polymorphism have been previously associated to decreased NLRC4 transcription and low IL-18 serum level, indicated once more that less activation of inflammasome and IL-18 production are beneficial for MS patients. PBM analysis showed that MS cells express higher level of inflammasome genes than HD ones, and are more prone to respond to a classical NLRP3 stimulus than HD.
Collapse
Affiliation(s)
- Jaine Ls Soares
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomédicas (ICB), Universidade de Sao Paulo (USP). Avenida Professor Lineu Prestes 1730, Sao Paulo, Brazil
| | - Enedina Ml Oliveira
- Ambulatório de Doenças Desmielinizantes, Departamento de Neurologia e Neurocirurgia, Universidade Federal de Sao Paulo (UNIFESP). Rua Pedro de Toledo 650, Sao Paulo, Brazil
| | - Alessandra Pontillo
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomédicas (ICB), Universidade de Sao Paulo (USP). Avenida Professor Lineu Prestes 1730, Sao Paulo, Brazil.
| |
Collapse
|
14
|
Rea IM, Gibson DS, McGilligan V, McNerlan SE, Alexander HD, Ross OA. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front Immunol 2018; 9:586. [PMID: 29686666 PMCID: PMC5900450 DOI: 10.3389/fimmu.2018.00586] [Citation(s) in RCA: 817] [Impact Index Per Article: 116.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 03/08/2018] [Indexed: 12/11/2022] Open
Abstract
Cytokine dysregulation is believed to play a key role in the remodeling of the immune system at older age, with evidence pointing to an inability to fine-control systemic inflammation, which seems to be a marker of unsuccessful aging. This reshaping of cytokine expression pattern, with a progressive tendency toward a pro-inflammatory phenotype has been called "inflamm-aging." Despite research there is no clear understanding about the causes of "inflamm-aging" that underpin most major age-related diseases, including atherosclerosis, diabetes, Alzheimer's disease, rheumatoid arthritis, cancer, and aging itself. While inflammation is part of the normal repair response for healing, and essential in keeping us safe from bacterial and viral infections and noxious environmental agents, not all inflammation is good. When inflammation becomes prolonged and persists, it can become damaging and destructive. Several common molecular pathways have been identified that are associated with both aging and low-grade inflammation. The age-related change in redox balance, the increase in age-related senescent cells, the senescence-associated secretory phenotype (SASP) and the decline in effective autophagy that can trigger the inflammasome, suggest that it may be possible to delay age-related diseases and aging itself by suppressing pro-inflammatory molecular mechanisms or improving the timely resolution of inflammation. Conversely there may be learning from molecular or genetic pathways from long-lived cohorts who exemplify good quality aging. Here, we will discuss some of the current ideas and highlight molecular pathways that appear to contribute to the immune imbalance and the cytokine dysregulation, which is associated with "inflammageing" or parainflammation. Evidence of these findings will be drawn from research in cardiovascular disease, cancer, neurological inflammation and rheumatoid arthritis.
Collapse
Affiliation(s)
- Irene Maeve Rea
- School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, United Kingdom
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
- Care of Elderly Medicine, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - David S. Gibson
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Victoria McGilligan
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Susan E. McNerlan
- Regional Genetics Service, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - H. Denis Alexander
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, United States
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Vergara C, Thio C, Latanich R, Cox AL, Kirk GD, Mehta SH, Busch M, Murphy EL, Villacres MC, Peters MG, French AL, Golub E, Eron J, Lahiri CD, Shrestha S, Gustafson D, Young M, Anastos K, Aouizerat B, Kim AY, Lauer G, Thomas DL, Duggal P. Genetic basis for variation in plasma IL-18 levels in persons with chronic hepatitis C virus and human immunodeficiency virus-1 infections. Genes Immun 2017; 18:82-87. [PMID: 28300059 PMCID: PMC5408324 DOI: 10.1038/gene.2017.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 12/27/2022]
Abstract
Inflammasomes are multi-protein complexes integrating pathogen-triggered signaling leading to the generation of pro-inflammatory cytokines, including interleukin-18 (IL-18). Hepatitis C virus (HCV) and human immunodeficiency virus-1 (HIV) infections are associated with elevated IL-18, suggesting inflammasome activation. However, there is marked person-to-person variation in the inflammasome response to HCV and HIV. We hypothesized that host genetics may explain this variation. To test this, we analyzed the associations of plasma IL-18 levels and polymorphisms in 10 genes in the inflammasome cascade. 1538 participants with active HIV and/or HCV infection in 3 ancestry groups are included. Samples were genotyped using the Illumina Omni 1-quad and Omni 2.5 arrays. Linear regression analyses were performed to test the association of variants with logIL-18 including HCV and HIV infection status and HIV-RNA, in each ancestry group and then meta-analyzed. Eleven highly correlated SNPs (r2=0.98-1) in the IL18-BCO2 region were significantly associated with logIL-18; Each T allele of rs80011693 confers a decrease of 0.06 log pg/mL of IL-18 after adjusting for covariates (rs80011693; rs111311302 β=-0.06, P-value=2.7×10-4). In conclusion, genetic variation in IL18 is associated with IL-18 production in response to HIV and HCV infection and may explain variability in the inflammatory outcomes of chronic viral infections.
Collapse
Affiliation(s)
- C Vergara
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - C Thio
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - R Latanich
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - A L Cox
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - G D Kirk
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - S H Mehta
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - M Busch
- University of California, San Francisco, CA, USA
| | - E L Murphy
- University of California, San Francisco, CA, USA.,Blood Systems Research Institute, San Francisco, CA, USA
| | - M C Villacres
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - M G Peters
- Blood Systems Research Institute, San Francisco, CA, USA
| | - A L French
- CORE Center/Stroger Hospital of Cook County, Chicago, IL, USA
| | - E Golub
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - J Eron
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - C D Lahiri
- School of Medicine, Emory University, Atlanta, GA, USA
| | - S Shrestha
- The University of Alabama at Birmingham, AL, USA
| | - D Gustafson
- State University of New York-Downstate Medical Center, New York, NY, USA
| | - M Young
- Georgetown University Medical Center, Washington, DC, USA
| | - K Anastos
- Albert Einstein College of Medicine and Montefiore Medical Center, New York, NY, USA
| | - B Aouizerat
- Bluestone Center for Clinical Research, New York University, New York, NY, USA.,Department of Oral and Maxillofacial Surgery, New York University, New York, NY, USA
| | - A Y Kim
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - G Lauer
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - D L Thomas
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - P Duggal
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
Foebel AD, Pedersen NL. Genetic Influences on Functional Capacities in Aging. THE GERONTOLOGIST 2017; 56 Suppl 2:S218-29. [PMID: 26994262 DOI: 10.1093/geront/gnw006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Older populations are characterized by great heterogeneity in functional capacities and understanding the factors underlying these differences has been a major area of research for some decades. Genetic differences arguably play an important role in the heterogeneity observed for many outcomes among older individuals. However, the role of genes in the variation and trajectories of functional capacities in older age is poorly understood. This review was conducted to explore the evidence for genetic influences on physical functional capacities in aging. DESIGN AND METHODS This rapid review was conducted using the following criteria: journal articles retrieved from the PubMed, Embase, AgeLine, Scopus, and Web of Science electronic databases including the key words: genetics, genotype, polymorphism, physical or functional performance, functional capacity, activities of daily living, older, and elderly. In total, 118 articles were included for initial review. RESULTS The heritability of objective measures of physical function ranges from 30% to 60% in studies of older twins. There is a paucity of evidence about genetic influences on functional capacities, but some candidate genes related to functional capacity have been identified. IMPLICATIONS No strong candidate genes exist for functional capacities. Current methodologies are beginning to generate new evidence about genetic influences on overall physical function at older ages, but the variety of measures of functional capacity makes evidence difficult to compare.
Collapse
Affiliation(s)
- Andrea D Foebel
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
da Silva WC, Oshiro TM, de Sá DC, Franco DDGS, Festa Neto C, Pontillo A. Genotyping and differential expression analysis of inflammasome genes in sporadic malignant melanoma reveal novel contribution of CARD8, IL1B and IL18 in melanoma susceptibility and progression. Cancer Genet 2016; 209:474-480. [PMID: 27810076 DOI: 10.1016/j.cancergen.2016.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/30/2016] [Accepted: 09/08/2016] [Indexed: 10/21/2022]
Abstract
Sporadic melanoma malignancy is correlated with constitutive secretion of IL-1β in transformed melanocytes suggesting the involvement of inflammasome in melanoma. Common variants in inflammasome genes are known to affect IL-1β expression. To investigate the contribution of inflammasome genetics in melanoma development and progression and to identify a potential prognostic marker, the distribution of selected inflammasome SNPs was analysed in a Brazilian case/control cohort of sporadic malignant melanoma (SMM) and then the expression of inflammasome components was evaluated in melanoma biopsies. Allele and gene-specific Taqman assays were implied for genotyping of case/control DNA samples and for relative expression analysis in skin biopsies respectively. CARD8 rs6509365 was found to be significantly more common in healthy volunteers than in SMM patients suggesting a protection effect of this variant towards melanoma development. Accordingly, CARD8 expression was found to be reduced in nevus compared to melanoma biopsies. Upon stratification, NLRP1 rs11651270 and CARD8 rs2043211 were found associated with nodular melanoma; IL1B rs1143643 to a lower value of Breslow index; IL18 rs5744256 to melanoma development in sun sensitive individuals. As expected, IL1B expression was up-regulated in tumour biopsies especially in metastatic samples, whereas IL18 was down-regulated compared to nevus. Our results demonstrated for the first time the contribution of inflammasome genes CARD8, IL1B and IL18 in SMM.
Collapse
Affiliation(s)
- Wanessa Cardoso da Silva
- Laboratory of Medical Investigation in Dermatology and Immunodeficiences-LIM 56, Department of Dermatology, Faculty of Medicine, University of Sao Paulo, Avenida Dr Eneas de Carvalho Aguiar, 470 - Prédio 2-3° andar, 05403-000 Cerqueira César, Sao Paulo, Brazil.
| | - Telma Miyuki Oshiro
- Laboratory of Medical Investigation in Dermatology and Immunodeficiences-LIM 56, Department of Dermatology, Faculty of Medicine, University of Sao Paulo, Avenida Dr Eneas de Carvalho Aguiar, 470 - Prédio 2-3° andar, 05403-000 Cerqueira César, Sao Paulo, Brazil
| | - Daniel Coelho de Sá
- Department of Dermatology, Faculty of Medicine, University of Sao Paulo, Avenida Dr. Eneas de Carvalho Aguiar 500, 05403-000 Cerqueira Cesar, Sao Paulo, Brazil
| | - Dilcilea D G S Franco
- Department of Dermatology, Faculty of Medicine, University of Sao Paulo, Avenida Dr. Eneas de Carvalho Aguiar 500, 05403-000 Cerqueira Cesar, Sao Paulo, Brazil
| | - Cyro Festa Neto
- Department of Dermatology, Faculty of Medicine, University of Sao Paulo, Avenida Dr. Eneas de Carvalho Aguiar 500, 05403-000 Cerqueira Cesar, Sao Paulo, Brazil
| | - Alessandra Pontillo
- Laboratory of Immunogenetics, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Avenida Prof. Lineu Prestes 1730, 05508-000 Cidade Universitaria, Sao Paulo, Brazil
| |
Collapse
|
18
|
Contribution of inflammasome genetics in Plasmodium vivax malaria. INFECTION GENETICS AND EVOLUTION 2016; 40:162-166. [DOI: 10.1016/j.meegid.2016.02.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/26/2016] [Accepted: 02/28/2016] [Indexed: 01/25/2023]
|
19
|
Mekli K, Marshall A, Nazroo J, Vanhoutte B, Pendleton N. Genetic variant of Interleukin-18 gene is associated with the Frailty Index in the English Longitudinal Study of Ageing. Age Ageing 2015; 44:938-42. [PMID: 26396182 PMCID: PMC4621230 DOI: 10.1093/ageing/afv122] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 06/17/2015] [Indexed: 01/30/2023] Open
Abstract
Background: the term frailty refers to a condition of increased vulnerability to stressors among older people, leading to a decline in homeostatic reserve. Frailty often leads to falls, hospitalisation and mortality, hence its importance for the delivery of health care to older adults. The pathophysiological mechanisms behind frailty are not well understood, but the decreased steroid-hormone production and elevated chronic systemic inflammation of older people appear to be major contributors. Method: we used a sample of 3,160 individuals aged 50 or older from the English Longitudinal Study of Ageing and assessed their frailty status according to a Frailty Index. We selected 620 single nucleotide polymorphisms in genes involved in the steroid hormone or inflammatory pathways. We performed linear association analysis. The outcome variable was the square root transformation of the Frailty Index, with age and sex entered as covariates. Results: the strongest signal was detected in the pro-inflammatory Interleukin-18 gene (rs360722, P = 0.0021, β = −0.015). Further significant signals were observed in the Interleukin-12 (rs4679868, P = 0.0062, β = −0.008 and rs9852519, P = 0.0077, β = −0.008), low density lipoprotein receptor-related protein 1 (rs1799986, P = 0.0065, β = 0.011) and Selectin-P (rs6131, P = 0.0097, β = −0.01) genes. None of these associations remain significant after Bonferroni correction. Conclusions: we show potential associations between genetic variants of four genes and the frailty index. These genes are involved in the cholesterol transport and inflammatory pathway and, as such, our results provide further support for the involvement of the immunological processes in frailty of the elderly.
Collapse
Affiliation(s)
- Krisztina Mekli
- Cathie Marsh Institute for Social Research, University of Manchester, Manchester M13 9PT, UK
| | - Alan Marshall
- Cathie Marsh Institute for Social Research, University of Manchester, Manchester M13 9PT, UK present address: School of Geography and Geosciences, University of St Andrews, St Andrews, UK
| | - James Nazroo
- Cathie Marsh Institute for Social Research, University of Manchester, Manchester M13 9PT, UK
| | - Bram Vanhoutte
- Cathie Marsh Institute for Social Research, University of Manchester, Manchester M13 9PT, UK
| | - Neil Pendleton
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| |
Collapse
|
20
|
Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207229 DOI: 10.3978/j.issn.2305-5839.2015.03.49] [Citation(s) in RCA: 536] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the brain, which is characterized by the formation of extracellular amyloid plaques (or senile plaques) and intracellular neurofibrillary tangles. However, increasing evidences demonstrated that neuroinflammatory changes, including chronic microgliosis are key pathological components of AD. Microglia, the resident immune cells of the brain, is constantly survey the microenvironment under physiological conditions. In AD, deposition of β-amyliod (Aβ) peptide initiates a spectrum of cerebral neuroinflammation mediated by activating microglia. Activated microglia may play a potentially detrimental role by eliciting the expression of pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) influencing the surrounding brain tissue. Emerging studies have demonstrated that up-regulation of pro-inflammatory cytokines play multiple roles in both neurodegeneration and neuroprotection. Understanding the pro-inflammatory cytokines signaling pathways involved in the regulation of AD is crucial to the development of strategies for therapy. This review will discuss the mechanisms and important role of pro-inflammatory cytokines in the pathogenesis of AD, and the ongoing drug targeting pro-inflammatory cytokine for therapeutic modulation.
Collapse
Affiliation(s)
- Wen-Ying Wang
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Meng-Shan Tan
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jin-Tai Yu
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Lan Tan
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
21
|
Ethnicity-stratified analysis of the association between IL-18 polymorphisms and systemic lupus erythematosus in a European population: a meta-analysis. Arch Dermatol Res 2015; 307:747-55. [PMID: 26026656 DOI: 10.1007/s00403-015-1580-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 04/10/2015] [Accepted: 05/17/2015] [Indexed: 10/23/2022]
Abstract
We performed a meta-analysis to identify the association between polymorphisms in the promoter of interleukin-18 (IL-18) and susceptibility for systemic lupus erythematosus (SLE) . Genotype data for three single-nucleotide polymorphisms (SNPs rs360719, rs1946518, and rs187238) in the IL-18 promoter were extracted from 20 studies of three different ethnicities (European, Asian, and South American). Data from each ethnicity group and their combinations were analyzed. We found distinct evidence of an association between rs360719 and SLE (P = 0.001) in the European/South American group [odds ratio (OR) 1.31 per C allele, 95% confidence interval (CI) 1.11-1.53]. Stratification analysis by ethnicity showed a significant association between rs360719 and SLE in the European population (OR 1.33 per C allele, 95% CI 1.11-1.61, P = 0.003) and a lesser effect in the same direction in the South American population (OR 1.18). A significant association was also identified between rs1946518 and SLE in the European population (OR 1.16 per A allele, 95% CI 1.03-1.30, P = 0.017), although there was no association in the Asian or the combined European/Asian population. We also examined genome-wide association study (GWAS) data from an Asian subpopulation (Chinese) for the association between rs1946518 and SLE, but found no association (P = 0.83). The third SNP, rs187238, was not significantly associated with SLE in any of the populations examined. In summary, this study identified a significant association between SLE and two SNPs within the IL-18 gene promoter region (rs360719 and rs1946518) in a European population, but not in populations of Asian origin.
Collapse
|
22
|
Boef AGC, Dekkers OM, le Cessie S. Mendelian randomization studies: a review of the approaches used and the quality of reporting. Int J Epidemiol 2015; 44:496-511. [PMID: 25953784 DOI: 10.1093/ije/dyv071] [Citation(s) in RCA: 369] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Mendelian randomization (MR) studies investigate the effect of genetic variation in levels of an exposure on an outcome, thereby using genetic variation as an instrumental variable (IV). We provide a meta-epidemiological overview of the methodological approaches used in MR studies, and evaluate the discussion of MR assumptions and reporting of statistical methods. METHODS We searched PubMed, Medline, Embase and Web of Science for MR studies up to December 2013. We assessed (i) the MR approach used; (ii) whether the plausibility of MR assumptions was discussed; and (iii) whether the statistical methods used were reported adequately. RESULTS Of 99 studies using data from one study population, 32 used genetic information as a proxy for the exposure without further estimation, 44 performed a formal IV analysis, 7 compared the observed with the expected genotype-outcome association, and 1 used both the latter two approaches. The 80 studies using data from multiple study populations used many different approaches to combine the data; 52 of these studies used some form of IV analysis; 44% of studies discussed the plausibility of all three MR assumptions in their study. Statistical methods used for IV analysis were insufficiently described in 14% of studies. CONCLUSIONS Most MR studies either use the genotype as a proxy for exposure without further estimation or perform an IV analysis. The discussion of underlying assumptions and reporting of statistical methods for IV analysis are frequently insufficient. Studies using data from multiple study populations are further complicated by the combination of data or estimates. We provide a checklist for the reporting of MR studies.
Collapse
Affiliation(s)
- Anna G C Boef
- Department of Clinical Epidemiology, Department of Endocrinology and Metabolic Diseases, and Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, The Netherlands.
| | - Olaf M Dekkers
- Department of Clinical Epidemiology, Department of Endocrinology and Metabolic Diseases, and Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, The Netherlands. Department of Clinical Epidemiology, Department of Endocrinology and Metabolic Diseases, and Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Saskia le Cessie
- Department of Clinical Epidemiology, Department of Endocrinology and Metabolic Diseases, and Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, The Netherlands. Department of Clinical Epidemiology, Department of Endocrinology and Metabolic Diseases, and Department of Medical Statistics and Bioinformatics, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
23
|
At the interface of sensory and motor dysfunctions and Alzheimer's disease. Alzheimers Dement 2015; 11:70-98. [PMID: 25022540 PMCID: PMC4287457 DOI: 10.1016/j.jalz.2014.04.514] [Citation(s) in RCA: 416] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/13/2014] [Accepted: 04/09/2014] [Indexed: 11/21/2022]
Abstract
Recent evidence indicates that sensory and motor changes may precede the cognitive symptoms of Alzheimer's disease (AD) by several years and may signify increased risk of developing AD. Traditionally, sensory and motor dysfunctions in aging and AD have been studied separately. To ascertain the evidence supporting the relationship between age-related changes in sensory and motor systems and the development of AD and to facilitate communication between several disciplines, the National Institute on Aging held an exploratory workshop titled "Sensory and Motor Dysfunctions in Aging and AD." The scientific sessions of the workshop focused on age-related and neuropathologic changes in the olfactory, visual, auditory, and motor systems, followed by extensive discussion and hypothesis generation related to the possible links among sensory, cognitive, and motor domains in aging and AD. Based on the data presented and discussed at this workshop, it is clear that sensory and motor regions of the central nervous system are affected by AD pathology and that interventions targeting amelioration of sensory-motor deficits in AD may enhance patient function as AD progresses.
Collapse
|
24
|
Matteini AM, Li J, Lange EM, Tanaka T, Lange LA, Tracy RP, Wang Y, Biggs ML, Arking DE, Fallin MD, Chakravarti A, Psaty BM, Bandinelli S, Ferrucci L, Reiner AP, Walston JD. Novel gene variants predict serum levels of the cytokines IL-18 and IL-1ra in older adults. Cytokine 2013; 65:10-6. [PMID: 24182552 DOI: 10.1016/j.cyto.2013.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 08/23/2013] [Accepted: 10/04/2013] [Indexed: 10/26/2022]
Abstract
Activation of inflammatory pathways measured by serum inflammatory markers such as interleukin-18 (IL-18) and interleukin-1 receptor antagonist (IL-1ra) is strongly associated with the progression of chronic disease states in older adults. Given that these serum cytokine levels are in part a heritable trait, genetic variation may predict increased serum levels. Using the Cardiovascular Health Study and InCHIANTI cohorts, a genome-wide association study was performed to identify genetic variants that influence IL-18 and IL-1ra serum levels among older adults. Multiple linear regression models characterized the association between each SNP and log-transformed cytokine values. Tests for multiple independent signals within statistically significant loci were performed using haplotype analysis and regression models conditional on lead SNP in each region. Multiple SNPs were associated with these cytokines with genome-wide significance, including SNPs in the IL-18-BCO gene region of chromosome 2 for IL-18 (top SNP rs2250417, P=1.9×10(-32)) and in the IL-1 gene family region of chromosome 2 for IL-1ra (rs6743376, P=2.3×10(-26)). Haplotype tests and conditional linear regression models showed evidence of multiple independent signals in these regions. Serum IL-18 levels were also associated with a region on chromosome 2 containing the NLRC4 gene (rs12989936, P=2.7×10(-19)). These data characterize multiple robust genetic signals that influence IL-18 and IL-1ra cytokine production. In particular, the signal for serum IL-18 located on chromosome two is novel and potentially important in inflammasome triggered chronic activation of inflammation in older adults. Replication in independent cohorts is an important next step, as well as molecular studies to better understand the role of NLRC4.
Collapse
Affiliation(s)
- A M Matteini
- Division of Geriatric Medicine, Johns Hopkins Medical Institution, Baltimore, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bossù P, Cutuli D, Palladino I, Caporali P, Angelucci F, Laricchiuta D, Gelfo F, De Bartolo P, Caltagirone C, Petrosini L. A single intraperitoneal injection of endotoxin in rats induces long-lasting modifications in behavior and brain protein levels of TNF-α and IL-18. J Neuroinflammation 2012; 9:101. [PMID: 22642744 PMCID: PMC3444884 DOI: 10.1186/1742-2094-9-101] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 05/29/2012] [Indexed: 11/10/2022] Open
Abstract
Background Systemic inflammation might cause neuronal damage and sustain neurodegenerative diseases and behavior impairment, with the participation of pro-inflammatory cytokines, like tumor necrosis factor (TNF)-α and interleukin (IL)-18. However, the potential contribution of these cytokines to behavioral impairment in the long-term period has not been fully investigated. Methods Wistar rats were treated with a single intraperitoneal injection of LPS (5 mg/kg) or vehicle. After 7 days and 10 months, the animal behavior was evaluated by testing specific cognitive functions, as mnesic, discriminative, and attentional functions, as well as anxiety levels. Contextually, TNF-α and IL-18 protein levels were measured by ELISA in defined brain regions (that is, frontal cortex, hippocampus, striatum, cerebellum, and hypothalamus). Results Behavioral testing demonstrated a specific and persistent cognitive impairment characterized by marked deficits in reacting to environment modifications, possibly linked to reduced motivational or attentional deficits. Concomitantly, LPS induced a TNF-α increase in the hippocampus and frontal cortex (from 7 days onward) and cerebellum (only at 10 months). Interestingly, LPS treatment enhanced IL-18 expression in these same areas only at 10 months after injection. Conclusions Overall, these results indicate that the chronic neuroinflammatory network elicited by systemic inflammation involves a persistent participation of TNF-α accompanied by a differently regulated contribution of IL-18. This leads to speculation that, though with still unclear mechanisms, both cytokines might take part in long-lasting modifications of brain functions, including behavioral alteration.
Collapse
Affiliation(s)
- Paola Bossù
- Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, Via Ardeatina 30600179, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Basile G, Paffumi I, D'Angelo AG, Figliomeni P, Cucinotta MD, Pace E, Ferraro M, Saitta S, Lasco A, Gangemi S. Healthy centenarians show high levels of circulating interleukin-22 (IL-22). Arch Gerontol Geriatr 2012; 54:459-61. [PMID: 21640395 DOI: 10.1016/j.archger.2011.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 04/28/2011] [Accepted: 05/01/2011] [Indexed: 01/05/2023]
Abstract
Aging is characterized by a progressive alteration of homeostatic mechanisms modulated by environmental and genetic factors. It is associated with a pro-inflammatory status. In centenarians, an increase of pro-inflammatory cytokine production balanced by anti-inflammatory immune response that would promote longevity is observed. Cytokine dysregulation is believed to play a key role in the proposed remodeling of the immune-inflammatory responses accompanying old age. IL-22 is a pro-inflammatory cytokine belonging to the IL-10 family and represents an important effector molecule of activated T helper (Th)-22, Th-1, and Th-17 cells. We recruited 17 healthy centenarians (4 males, 13 females, range 100-105 years). All ultralongeval subjects were living at home or in a nursing home. Sixteen healthy, sex-matched individuals (4 males, 12 females, range 60-95 years) were also recruited as controls. Centenarians displayed significantly higher circulating IL-22 levels compared to control population (45.7±66.9 pg/ml versus 11.1±6.5 pg/ml; p=0.031). It's well known that IL-22 is a pro-inflammatory cytokine produced by activated T lymphocytes and NK cells. IL-22 stimulates the production of acute phase reactants and promotes the antimicrobial defense. The results of the present study show, for the first time, that there is an increase of IL-22 in healthy centenarians. This pro-inflammatory condition probably is protective against infection, promoting the longevity of these subjects.
Collapse
Affiliation(s)
- Giorgio Basile
- School and Unit of Geriatrics, Department of Internal Medicine, University of Messina, AOU Policlinico, Via C. Valeria n. 1, 98125 Messina, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Urbanek M, Hayes MG, Lee H, Freathy RM, Lowe LP, Ackerman C, Jafari N, Dyer AR, Cox NJ, Dunger DB, Hattersley AT, Metzger BE, Lowe WL. The role of inflammatory pathway genetic variation on maternal metabolic phenotypes during pregnancy. PLoS One 2012; 7:e32958. [PMID: 22479352 PMCID: PMC3316547 DOI: 10.1371/journal.pone.0032958] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 02/08/2012] [Indexed: 02/06/2023] Open
Abstract
Background Since mediators of inflammation are associated with insulin resistance, and the risk of developing diabetes mellitus and gestational diabetes, we hypothesized that genetic variation in members of the inflammatory gene pathway impact glucose levels and related phenotypes in pregnancy. We evaluated this hypothesis by testing for association between genetic variants in 31 inflammatory pathway genes in the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) cohort, a large multiethnic multicenter study designed to address the impact of glycemia less than overt diabetes on pregnancy outcome. Results Fasting, 1-hour, and 2-hour glucose, fasting and 1-hour C-peptide, and HbA1c levels were measured in blood samples obtained from HAPO participants during an oral glucose tolerance test at 24-32 weeks gestation. We tested for association between 458 SNPs mapping to 31 genes in the inflammatory pathway and metabolic phenotypes in 3836 European ancestry and 1713 Thai pregnant women. The strongest evidence for association was observed with TNF alpha and HbA1c (rs1052248; 0.04% increase per allele C; p-value = 4.4×10−5), RETN and fasting plasma glucose (rs1423096; 0.7 mg/dl decrease per allele A; p-value = 1.1×10−4), IL8 and 1 hr plasma glucose (rs2886920; 2.6 mg/dl decrease per allele T; p-value = 1.3×10−4), ADIPOR2 and fasting C-peptide (rs2041139; 0.55 ug/L decrease per allele A; p-value = 1.4×10−4), LEPR and 1-hour C-peptide (rs1171278; 0.62 ug/L decrease per allele T; p-value = 2.4×10−4), and IL6 and 1-hour plasma glucose (rs6954897; −2.29 mg/dl decrease per allele G, p-value = 4.3×10−4). Conclusions Based on the genes surveyed in this study the inflammatory pathway is unlikely to have a strong impact on maternal metabolic phenotypes in pregnancy although variation in individual members of the pathway (e.g. RETN, IL8, ADIPOR2, LEPR, IL6, and TNF alpha,) may contribute to metabolic phenotypes in pregnant women.
Collapse
Affiliation(s)
- Margrit Urbanek
- Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Saleh NM, Raj SM, Smyth DJ, Wallace C, Howson JMM, Bell L, Walker NM, Stevens HE, Todd JA. Genetic association analyses of atopic illness and proinflammatory cytokine genes with type 1 diabetes. Diabetes Metab Res Rev 2011; 27:838-43. [PMID: 22069270 PMCID: PMC3816329 DOI: 10.1002/dmrr.1259] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 06/03/2011] [Accepted: 06/08/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND The genetic basis of the autoimmune disease type 1 diabetes (T1D) has now been largely determined, so now we can compare these findings with emerging genetic knowledge of disorders and phenotypes that have been negatively or positively associated with T1D historically. Here, we assessed the role in T1D of variants previously reported to be associated with atopic diseases and epithelial barrier function, profilaggrin (FLG), and those that affect the expression levels of the proinflammatory cytokines tumour necrosis factor (TNF)-α, interleukin (IL)-1β, interferon (IFN)γ and IL-18. METHODS We genotyped single nucleotide polymorphisms (SNPs): -105/rs28665122 in SELS or SEPS1 (selenoprotein), three single nucleotide polymorphisms in IL18 (-105/rs360717, +183/rs5744292 and +1467/rs574456) and R501X/rs61816761 in FLG, the major locus associated with atopic dermatitis and predisposing to asthma, in a minimum of 6743 T1D cases and 7864 controls. RESULTS No evidence of T1D association was found for any of the SNPs we genotyped at FLG, SELS or IL18 (p≥0.03), nor with haplotypes of IL18 (p=0.82). Review of previous T1D genome-wide association results revealed that four (human leucocyte antigen (HLA), gasdermin B/ORM1 (Saccharomyces cerevisiae)-like/gasdermin B/, GSDMB/ORMDL3/GSDMA and IL2RB) of ten loci recently reported to be associated with asthma were associated with T1D (p≤0.005). CONCLUSIONS These results show that there are shared genetic associations for atopy-related traits and T1D, and this might help in the future to understand the mechanisms, pathways and environmental factors that underpin the rapid rise in incidence of both disorders in children.
Collapse
Affiliation(s)
- Nada M Saleh
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - Srilakshmi M Raj
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - Deborah J Smyth
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - Chris Wallace
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - Joanna M M Howson
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - Louise Bell
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - Neil M Walker
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - Helen E Stevens
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| | - John A Todd
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's HospitalCambridge CB2 0XY, UK
| |
Collapse
|
29
|
Guerra SG, Morris DL, Gateva V, Graham RR, Vyse TJ, Cunninghame Graham DS. Dense mapping of IL18 shows no association in SLE. Hum Mol Genet 2011; 20:1026-33. [PMID: 21149337 PMCID: PMC3033184 DOI: 10.1093/hmg/ddq536] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 11/30/2010] [Accepted: 12/06/2010] [Indexed: 01/27/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease which behaves as a complex genetic trait. At least 20 SLE risk susceptibility loci have been mapped using both candidate gene and genome-wide association strategies. The gene encoding the pro-inflammatory cytokine, IL18, has been reported as a candidate gene showing an association with SLE. This pleiotropic cytokine is expressed in a range of immune cells and has been shown to induce interferon-γ and tumour necrosis factor-α. Serum interleukin-18 has been reported to be elevated in patients with SLE. Here we aimed to densely map single nucleotide polymorphisms (SNPs) across IL18 to investigate the association across this locus. We genotyped 36 across IL18 by Illumina bead express in 372 UK SLE trios. We also genotyped these SNPs in a further 508 non-trio UK cases and were able to accurately impute a dense marker set across IL18 in WTCCC2 controls with a total of 258 SNPs. To improve the study's power, we also imputed a total of 158 SNPs across the IL18 locus using data from an SLE genome-wide association study and performed association testing. In total, we analysed 1818 cases and 10 770 controls in this study. Our large well-powered study (98% to detect odds ratio = 1.5, with respect to rs360719) showed that no individual SNP or haplotype was associated with SLE in any of the cohorts studied. We conclude that we were unable to replicate the SLE association with rs360719 located upstream of IL18. No evidence for association with any other common variant at IL18 with SLE was found.
Collapse
Affiliation(s)
| | - David L. Morris
- Rheumatology Section, Imperial College, London W12 0NN, UK and
| | - Vesela Gateva
- Immunology Biomarkers Group, Genentech, South San Francisco, CA, USA
| | - Robert R. Graham
- Immunology Biomarkers Group, Genentech, South San Francisco, CA, USA
| | - Timothy J. Vyse
- Rheumatology Section, Imperial College, London W12 0NN, UK and
| | | |
Collapse
|
30
|
Zhang XM, Jin T, Quezada HC, Mix E, Winblad B, Zhu J. Kainic acid-induced microglial activation is attenuated in aged interleukin-18 deficient mice. J Neuroinflammation 2010; 7:26. [PMID: 20398244 PMCID: PMC2865455 DOI: 10.1186/1742-2094-7-26] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 04/14/2010] [Indexed: 12/28/2022] Open
Abstract
Background Previously, we found that interleukin (IL)-18 deficiency aggravates kainic acid (KA)-induced hippocampal neurodegeneration in young C57BL/6 mice due to an over-compensation by IL-12. Additionally, IL-18 participates in fundamental inflammatory processes that increase during aging. In the present study, we were interested in the role of IL-18 in KA-induced neurodegeneration in aged female C57BL/6 mice. Methods Fifteen aged female IL-18 knockout (KO) and 15 age-matched wild-type (WT) mice (18 to 19 months old) were treated with KA at a dose of 25 mg/kg body weight intranasally. Seizure activities and behavioral changes were rated using a 6-point scoring system and open-field test, respectively. Seven days after KA treatment, degenerating neurons were detected by Nissl's method and Fluoro-Jade B staining; and microglial activation was analyzed by immunohistochemistry and flow cytometry. Results Aged female IL-18 KO and WT mice showed similar responses to treatment with KA as demonstrated by comparable seizure activities, behavioral changes and neuronal cell death. However, aged female IL-18 KO mice failed to exhibit the strong microglial activation shown in WT mice. Interestingly, even though the number of activated microglia was less in KA-treated IL-18 KO mice than in KA-treated WT mice, the proportion of microglia that expressed the cytokines tumor necrosis factor (TNF)-α, IL-6 and IL-10 was higher in KA-treated IL-18 KO mice. Conclusion Deficiency of IL-18 attenuates microglial activation after KA-induced excitotoxicity in aged brain, while the net effects of IL-18 deficiency are balanced by the enhancement of other cytokines, such as TNF-α, IL-6 and IL-10.
Collapse
Affiliation(s)
- Xing-Mei Zhang
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
31
|
He M, Cornelis MC, Kraft P, van Dam RM, Sun Q, Laurie CC, Mirel DB, Chasman DI, Ridker PM, Hunter DJ, Hu FB, Qi L. Genome-wide association study identifies variants at the IL18-BCO2 locus associated with interleukin-18 levels. Arterioscler Thromb Vasc Biol 2010; 30:885-90. [PMID: 20150558 PMCID: PMC2841960 DOI: 10.1161/atvbaha.109.199422] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Interleukin-18 (IL-18) is a proinflammatory cytokine involved in the processes of innate and acquired immunities and associated with cardiovascular disease and type 2 diabetes. We sought to identify the common genetic variants associated with IL-18 levels. METHODS AND RESULTS We performed a 2-stage genome-wide association study among women of European ancestry from the Nurses' Health Study (NHS) and Women's Genome Health Study (WGHS). IL-18 levels were measured by ELISA. In the discovery stage (NHS, n=1523), 7 single-nucleotide polymorphisms (SNPs) at the IL18-BCO2 locus were associated with IL-18 concentrations at the 1 x 10(-5) significance level. The strongest association was found for SNP rs2115763 in the BCO2 gene (P=6.31 x 10(-8)). In silico replication in WGHS (435 women) confirmed these findings. The combined analysis of the 2 studies indicated that SNPs rs2115763, rs1834481, and rs7106524 reached a genome-wide significance level (P<5 x 10(-8)). Forward selection analysis indicated that SNPs rs2115763 and rs1834481 were independently associated with IL-18 levels (P=0.0002 and 0.0006, respectively). The 2 SNPs together explained 2.9% of variation of plasma IL-18 levels. CONCLUSIONS This study identified several novel variants at the IL18-BCO2 locus associated with IL-18 levels.
Collapse
Affiliation(s)
- Meian He
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Institute of Occupational Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Marilyn C. Cornelis
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
| | - Rob M. van Dam
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Qi Sun
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
| | - Cathy C. Laurie
- Department of Biostatistics, University of Washington, Seattle 98195, WA, USA
| | - Daniel B. Mirel
- Center for Genotyping and Analysis, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge 02142, MA,USA
| | - Daniel I. Chasman
- Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
- Donald W. Reynolds Center for Cardiovascular Research, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
| | - Paul M. Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
- Donald W. Reynolds Center for Cardiovascular Research, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Frank B. Hu
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| |
Collapse
|
32
|
Bochud M, Rousson V. Usefulness of Mendelian randomization in observational epidemiology. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2010; 7:711-28. [PMID: 20616999 PMCID: PMC2872313 DOI: 10.3390/ijerph7030711] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Accepted: 02/16/2010] [Indexed: 11/16/2022]
Abstract
Mendelian randomization refers to the random allocation of alleles at the time of gamete formation. In observational epidemiology, this refers to the use of genetic variants to estimate a causal effect between a modifiable risk factor and an outcome of interest. In this review, we recall the principles of a "Mendelian randomization" approach in observational epidemiology, which is based on the technique of instrumental variables; we provide simulations and an example based on real data to demonstrate its implications; we present the results of a systematic search on original articles having used this approach; and we discuss some limitations of this approach in view of what has been found so far.
Collapse
Affiliation(s)
- Murielle Bochud
- University Institute of Social and Preventive Medicine, Rue du Bugnon 17, Lausanne, Switzerland.
| | | |
Collapse
|
33
|
Dato S, Krabbe KS, Thinggaard M, Pedersen BK, Christensen K, Bruunsgaard H, Christiansen L. Commonly studied polymorphisms in inflammatory cytokine genes show only minor effects on mortality and related risk factors in nonagenarians. J Gerontol A Biol Sci Med Sci 2010; 65:225-35. [PMID: 20083555 DOI: 10.1093/gerona/glp210] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Systemic low-grade inflammation is consistently associated with functional status, cognitive functioning, multimorbidity, and survival in oldest olds. If inflammation is either a cause or a consequence of age-related pathology, genetic determinants of late-life survival can reside in cytokine genes polymorphisms, regulating inflammatory responses. The aim of this study was to test associations between commonly studied polymorphisms in interleukin (IL)6, IL10, IL15, and IL18, and tumor necrosis factor-alpha genes and late-life survival in a longitudinal cohort of nonagenarians: the Danish 1905 cohort. Additionally, associations were investigated between inflammatory markers and major predictors of mortality as cognitive and functional status. Modest sex-specific associations were found with survival, cognitive functioning, and handgrip strength. Evaluation of combined genotypes indicated that, in nonagenarian men, the balance of pro- and anti-inflammatory activity at IL18 and IL10 loci is protective against cognitive decline. In conclusion, in this large study with virtually complete follow-up, commonly studied polymorphisms in cytokine genes do not have a major impact on late-life survival or associated risk phenotypes.
Collapse
Affiliation(s)
- Serena Dato
- The Danish Aging Research Center, Epidemiology Unit, Institute of Public Health, University of Southern Denmark, J.B. Winsløws Vej 9, DK-5000 Odense, Denmark.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Nutritional epidemiology aims to identify dietary and lifestyle causes for human diseases. Causality inference in nutritional epidemiology is largely based on evidence from studies of observational design, and may be distorted by unmeasured or residual confounding and reverse causation. Mendelian randomization is a recently developed methodology that combines genetic and classical epidemiological analysis to infer causality for environmental exposures, based on the principle of Mendel's law of independent assortment. Mendelian randomization uses genetic variants as proxies for environmental exposures of interest. Associations derived from Mendelian randomization analysis are less likely to be affected by confounding and reverse causation. During the past 5 years, a body of studies examined the causal effects of diet/lifestyle factors and biomarkers on a variety of diseases. The Mendelian randomization approach also holds considerable promise in the study of intrauterine influences on offspring health outcomes. However, the application of Mendelian randomization in nutritional epidemiology has some limitations.
Collapse
Affiliation(s)
- Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA.
| |
Collapse
|
35
|
Thomas K, Rafiq S, Frayling TM, Ebrahim S, Kumari M, Gallacher J, Ferrucci L, Bandinelli S, Wallace RB, Melzer D, Martin RM, Ben-Shlomo Y. Interleukin-18 polymorphism and physical functioning in older people: a replication study and meta-analysis. J Gerontol A Biol Sci Med Sci 2009; 64:1177-82. [PMID: 19633236 PMCID: PMC2981454 DOI: 10.1093/gerona/glp092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Accepted: 02/09/2009] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Levels of the proinflammatory cytokine interleukin-18 (IL-18) are raised in old age and are associated with reduced physical functioning. Previous studies have indicated that the C allele of the rs5744256 polymorphism in the IL-18 gene is strongly associated with reduced circulating IL-18 levels. This variant has previously been associated with improved locomotor performance in old age, but the finding requires independent replication. METHODS We examined the association between the IL-18 polymorphism rs5744256 and physical functioning in three cohorts with a total of 4,107 participants aged 60-85 years: the English Longitudinal Study of Ageing, Caerphilly, and Boyd Orr. We meta-analyzed (N = 6,141) the results with data from the original paper reporting this association: Iowa-Established Populations for Epidemiological Study of the Elderly and InCHIANTI cohorts. Physical functioning was assessed by timed walks or the get up and go test. As locomotor performance tests differed between the cohorts and the distributions of times to complete the test (in seconds) were positively skewed, we used the reciprocal transformation and computed study-specific z scores. RESULTS Based on the three new studies, the estimated linear regression coefficient per C allele was 0.011 (95% confidence interval [95% CI]: -0.04 to 0.06). A meta-analysis that pooled the data from all studies showed weak evidence of an effect, with a regression coefficient of 0.047 (95% CI: 0.010 to 0.083). CONCLUSIONS We did not replicate an association between the IL-18 rs5744256 polymorphism and the physical function in people aged 60-85 years. However, pooling data from all studies suggested a weak association of the C allele of the rs5744256 single nucleotide polymorphism on improving walking times in old age.
Collapse
Affiliation(s)
- Kate Thomas
- Department of Social Medicine, University of Bristol, Bristol BS8 2PS, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hollegaard MV, Grove J, Thorsen P, Nørgaard-Pedersen B, Hougaard DM. High-Throughput Genotyping on Archived Dried Blood Spot Samples. Genet Test Mol Biomarkers 2009; 13:173-9. [DOI: 10.1089/gtmb.2008.0073] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Mads V. Hollegaard
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
- NANEA, Department of Epidemiology, Institute of Public Health, University of Aarhus, Aarhus, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Jakob Grove
- NANEA, Department of Epidemiology, Institute of Public Health, University of Aarhus, Aarhus, Denmark
| | - Poul Thorsen
- NANEA, Department of Epidemiology, Institute of Public Health, University of Aarhus, Aarhus, Denmark
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Bent Nørgaard-Pedersen
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - David M. Hougaard
- Department of Clinical Biochemistry and Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
37
|
Smith AJP, Humphries SE. Cytokine and cytokine receptor gene polymorphisms and their functionality. Cytokine Growth Factor Rev 2009; 20:43-59. [PMID: 19038572 DOI: 10.1016/j.cytogfr.2008.11.006] [Citation(s) in RCA: 276] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cytokines, signaling proteins produced by a variety of cell types, are essential for the development and functioning of both innate and adaptive immune response. Cytokine gene expression is tightly regulated, and aberrant expression from environmental and genetic polymorphism has been implicated in a range of diseases, susceptibility to infections, and responses to treatment. This review concentrates on the functionality of cytokine and cytokine receptor gene polymorphisms; it is through these variants that genuine disease-associations are based. Several mechanisms for single nucleotide polymorphism (SNP) functionality are present within cytokine genes including: amino acid changes (IL-6R, IL-13, IL-1alpha), exon skipping (IL-7Ralpha), proximal promoter variants (IL-1beta, IL-Ra, IL-2, IL-6, IL-10, IL-12, IL-13, IL-16, TNF, IFN-gamma, TGF-beta), distal promoter variants (IL-6, IL-18) and intronic enhancer variants (IL-8).
Collapse
Affiliation(s)
- Andrew J P Smith
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, United Kingdom.
| | | |
Collapse
|
38
|
Rafiq S, Melzer D, Weedon MN, Lango H, Saxena R, Scott LJ, DIAGRAM Consortium, Palmer CNA, Morris AD, McCarthy MI, Ferrucci L, Hattersley AT, Zeggini E, Frayling TM. Gene variants influencing measures of inflammation or predisposing to autoimmune and inflammatory diseases are not associated with the risk of type 2 diabetes. Diabetologia 2008; 51:2205-13. [PMID: 18853133 PMCID: PMC2662689 DOI: 10.1007/s00125-008-1160-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 08/18/2008] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS There are strong associations between measures of inflammation and type 2 diabetes, but the causal directions of these associations are not known. We tested the hypothesis that common gene variants known to alter circulating levels of inflammatory proteins, or known to alter autoimmune-related disease risk, influence type 2 diabetes risk. METHODS We selected 46 variants: (1) eight variants known to alter circulating levels of inflammatory proteins, including those in the IL18, IL1RN, IL6R, MIF, PAI1 (also known as SERPINE1) and CRP genes; and (2) 38 variants known to predispose to autoimmune diseases, including type 1 diabetes. We tested the associations of these variants with type 2 diabetes using a meta-analysis of 4,107 cases and 5,187 controls from the Wellcome Trust Case Control Consortium, the Diabetes Genetics Initiative, and the Finland-United States Investigation of NIDDM studies. We followed up associated variants (p < 0.01) in a further set of 3,125 cases and 3,596 controls from the UK. RESULTS We found no evidence that inflammatory or autoimmune disease variants are associated with type 2 diabetes (at p CONCLUSIONS/INTERPRETATION Our study provided no evidence that variants known to alter measures of inflammation, autoimmune or inflammatory disease risk, including type 1 diabetes, alter type 2 diabetes risk.
Collapse
Affiliation(s)
- S Rafiq
- Peninsula Medical School, University of Exeter, Magdalen Road, Exeter, EX1 2LU, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Nuala Sheehan and colleagues describe how Mendelian randomization provides an alternative way of dealing with the problems of observational studies, especially confounding.
Collapse
Affiliation(s)
- Nuala A Sheehan
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom.
| | | | | | | |
Collapse
|
40
|
Liu W, Tang Q, Jiang H, Ding X, Liu Y, Zhu R, Tang Y, Li B, Wei M. Promoter polymorphism of interleukin-18 in angiographically proven coronary artery disease. Angiology 2008; 60:180-5. [PMID: 18599493 DOI: 10.1177/0003319708319939] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Interleukin 18 (IL-18) is a pro-atherogenic cytokine associated with the occurrence of various cardiac complications. The IL-18 gene has a functional -137 G/C polymorphism (rs187238) in the promoter region. Using the ligase detection reaction-polymerase chain reaction, we genotyped a cohort of patients in Chinese Han population in Xiangfan region. Case patients of coronary artery disease and control patients were identified by coronary angiography. The plasma IL-18 concentrations were measured by ELISA. A significant increase of G allele or GG-genotype was observed in 241 case patients compared to 145 control individuals (frequency of G allele = 0.90 vs 0.83, p=0.004; frequency of GG-genotype = 0.81 vs 0.68, p = 0.005). In case patients, G allele carriers in multi-vessel disease patients had a higher occurrence rate when compared to single-vessel disease patients, but no significant difference was detected (frequency of G allele = 0.92 vs 0.88, p=0.107; frequency of GG-genotype = 0.84 vs 0.75, p = 0.089). IL-18 protein concentration of the -137GG genotype was much higher than concentration of the CG and CC genotype (case patients: 229.1+/-131.5 vs 122.7+/-73.6 pg/ml, P < 0.001; control patients: 65.9+/-31.6 vs 42.4+/-19.5 pg/ml, P < 0.001). To conclude, IL-18 promoter -137G/C polymorphism influences IL-18 levels and the occurrence of coronary artery disease, suggesting that IL-18 is causally involved in the development of atherosclerosis.
Collapse
Affiliation(s)
- Wenwei Liu
- Department of Cardiology, Xiangfan Central Hospital, Xiangfan, Hubei, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Melzer D, Perry JRB, Hernandez D, Corsi AM, Stevens K, Rafferty I, Lauretani F, Murray A, Gibbs JR, Paolisso G, Rafiq S, Simon-Sanchez J, Lango H, Scholz S, Weedon MN, Arepalli S, Rice N, Washecka N, Hurst A, Britton A, Henley W, van de Leemput J, Li R, Newman AB, Tranah G, Harris T, Panicker V, Dayan C, Bennett A, McCarthy MI, Ruokonen A, Jarvelin MR, Guralnik J, Bandinelli S, Frayling TM, Singleton A, Ferrucci L. A genome-wide association study identifies protein quantitative trait loci (pQTLs). PLoS Genet 2008; 4:e1000072. [PMID: 18464913 PMCID: PMC2362067 DOI: 10.1371/journal.pgen.1000072] [Citation(s) in RCA: 398] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 04/11/2008] [Indexed: 11/18/2022] Open
Abstract
There is considerable evidence that human genetic variation influences gene expression. Genome-wide studies have revealed that mRNA levels are associated with genetic variation in or close to the gene coding for those mRNA transcripts – cis effects, and elsewhere in the genome – trans effects. The role of genetic variation in determining protein levels has not been systematically assessed. Using a genome-wide association approach we show that common genetic variation influences levels of clinically relevant proteins in human serum and plasma. We evaluated the role of 496,032 polymorphisms on levels of 42 proteins measured in 1200 fasting individuals from the population based InCHIANTI study. Proteins included insulin, several interleukins, adipokines, chemokines, and liver function markers that are implicated in many common diseases including metabolic, inflammatory, and infectious conditions. We identified eight Cis effects, including variants in or near the IL6R (p = 1.8×10−57), CCL4L1 (p = 3.9×10−21), IL18 (p = 6.8×10−13), LPA (p = 4.4×10−10), GGT1 (p = 1.5×10−7), SHBG (p = 3.1×10−7), CRP (p = 6.4×10−6) and IL1RN (p = 7.3×10−6) genes, all associated with their respective protein products with effect sizes ranging from 0.19 to 0.69 standard deviations per allele. Mechanisms implicated include altered rates of cleavage of bound to unbound soluble receptor (IL6R), altered secretion rates of different sized proteins (LPA), variation in gene copy number (CCL4L1) and altered transcription (GGT1). We identified one novel trans effect that was an association between ABO blood group and tumour necrosis factor alpha (TNF-alpha) levels (p = 6.8×10−40), but this finding was not present when TNF-alpha was measured using a different assay , or in a second study, suggesting an assay-specific association. Our results show that protein levels share some of the features of the genetics of gene expression. These include the presence of strong genetic effects in cis locations. The identification of protein quantitative trait loci (pQTLs) may be a powerful complementary method of improving our understanding of disease pathways. One of the central dogmas of molecular genetics is that DNA is transcribed to RNA which is translated to protein and alterations to proteins can influence human diseases. Genome-wide association studies have recently revealed many new DNA variants that influence human diseases. To complement these efforts, several genome-wide studies have established that DNA variation influences mRNA expression levels. Loci influencing mRNA levels have been termed “eQTLs”. In this study we have performed the first genome-wide association study of the third piece in this jigsaw – the role of DNA variation in relation to protein levels, or “pQTLs”. We analysed 42 proteins measured in blood fractions from the InCHIANTI study. We identified eight cis effects including common variants in or near the IL6R, CCL4, IL18, LPA, GGT1, SHBG, CRP and IL1RN genes, all associated with blood levels of their respective protein products. Mechanisms implicated included altered transcription (GGT1) but also rates of cleavage of bound to unbound soluble receptor (IL6R), altered secretion rates of different sized proteins (LPA) and variation in gene copy number (CCL4). Blood levels of many of these proteins are correlated with human diseases and the identification of “pQTLs” may in turn help our understanding of disease.
Collapse
Affiliation(s)
- David Melzer
- Department of Epidemiology and Public Health, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - John R. B. Perry
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Anna-Maria Corsi
- Tuscany Regional Health Agency, I.O.T. and Department of Medical and Surgical Critical Care, University of Florence, Florence, Italy
| | - Kara Stevens
- Department of Epidemiology and Public Health, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - Ian Rafferty
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Fulvio Lauretani
- Tuscany Regional Health Agency, I.O.T. and Department of Medical and Surgical Critical Care, University of Florence, Florence, Italy
| | - Anna Murray
- Department of Epidemiology and Public Health, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - J. Raphael Gibbs
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Giuseppe Paolisso
- Department of Geriatric Medicine and Metabolic Diseases, Second University of Naples, Naples, Italy
| | - Sajjad Rafiq
- Department of Epidemiology and Public Health, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - Javier Simon-Sanchez
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Hana Lango
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - Sonja Scholz
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Michael N. Weedon
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - Sampath Arepalli
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Neil Rice
- Department of Epidemiology and Public Health, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - Nicole Washecka
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Alison Hurst
- Department of Epidemiology and Public Health, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
| | - Angela Britton
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - William Henley
- School of Mathematics and Statistics, University of Plymouth, Plymouth, United Kingdom
| | - Joyce van de Leemput
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Rongling Li
- Department of Preventive Medicine and Center for Genomics and Bioinformatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Anne B. Newman
- University of Pittsburgh, Graduate School of Public Health, Departments of Epidemiology and Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Greg Tranah
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, United States of America
| | - Tamara Harris
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | - Vijay Panicker
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- Henry Wellcome Laboratories for Integrative Neurosciences and Endocrinology, University of Bristol, Bristol, United Kingdom
| | - Colin Dayan
- Henry Wellcome Laboratories for Integrative Neurosciences and Endocrinology, University of Bristol, Bristol, United Kingdom
| | - Amanda Bennett
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Headington, Oxford, United Kingdom
| | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Headington, Oxford, United Kingdom
- The Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, United Kingdom
| | - Aimo Ruokonen
- Department of Clinical Chemistry, University of Oulu, Oulu, Finland
| | - Marjo-Riitta Jarvelin
- Department of Public Health, Science, and General Practice, University of Oulu, Oulu, Finland
- Department of Epidemiology and Public Health, Imperial College London, London, United Kingdom
| | - Jack Guralnik
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, Bethesda, Maryland, United States of America
| | | | - Timothy M. Frayling
- Genetics of Complex Traits, Institute of Biomedical and Clinical Sciences, Peninsula College of Medicine and Dentistry, University of Exeter, Devon, United Kingdom
- * E-mail:
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute of Aging, Porter Neuroscience Research Center, Bethesda, Maryland, United States of America
| | - Luigi Ferrucci
- Longitudinal Studies Section, Clinical Research Branch, Gerontology Research Center, National Institute on Aging, Baltimore, Maryland, United States of America
| |
Collapse
|
42
|
Lawlor DA, Harbord RM, Sterne JAC, Timpson N, Davey Smith G. Mendelian randomization: using genes as instruments for making causal inferences in epidemiology. Stat Med 2008; 27:1133-63. [PMID: 17886233 DOI: 10.1002/sim.3034] [Citation(s) in RCA: 2937] [Impact Index Per Article: 172.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Observational epidemiological studies suffer from many potential biases, from confounding and from reverse causation, and this limits their ability to robustly identify causal associations. Several high-profile situations exist in which randomized controlled trials of precisely the same intervention that has been examined in observational studies have produced markedly different findings. In other observational sciences, the use of instrumental variable (IV) approaches has been one approach to strengthening causal inferences in non-experimental situations. The use of germline genetic variants that proxy for environmentally modifiable exposures as instruments for these exposures is one form of IV analysis that can be implemented within observational epidemiological studies. The method has been referred to as 'Mendelian randomization', and can be considered as analogous to randomized controlled trials. This paper outlines Mendelian randomization, draws parallels with IV methods, provides examples of implementation of the approach and discusses limitations of the approach and some methods for dealing with these.
Collapse
|
43
|
Huang MC, Liao JJ, Bonasera S, Longo DL, Goetzl EJ. Nuclear factor-kappaB-dependent reversal of aging-induced alterations in T cell cytokines. FASEB J 2008; 22:2142-50. [PMID: 18267981 DOI: 10.1096/fj.07-103721] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immunosenescence is characterized by decreases in protective immune responses and increases in inflammation and autoimmunity. The T helper (Th)17 subset of cluster-of-differentiation (CD)4 T cells, which is identified by its generation of interleukin (IL) -17, is implicated in autoimmune pathogenesis. To elucidate immunosenescent changes in Th17 cell cytokines, splenic CD4 T cells from 22- to 24-month-old (old) mice and 6- to 10-wk-old (young) mice were incubated on anti-CD3 plus anti-CD28 (anti-T cell antigen receptor) antibodies. After 96 h, T cells of old C57BL/6 and CBA mice generated up to 20-fold more IL-17 and up to 3-fold more IL-6 than those of young mice; T cells of young mice generated up to 5-fold more IL-21 than those of old mice; and no difference was found for IFN-gamma. At 24 h, cytokine mRNA levels paralleled 96 h cytokine concentrations. Naive CD4 T cells from old mice incubated on anti-T cell antigen receptor antibodies with transforming growth factor-beta, IL-1, IL-6, and IL-23 to induce de novo differentiation of Th17 cells had more IL-17 mRNA and produced more IL-17 than those of young mice. BAY11-7082 and the phytochemicals triptolide and butein suppressed nuclear concentrations of nuclear factor-kappaB and secreted levels of IL-17, IL-21, and IFN-gamma in parallel, with greater potency in Th17 cells from young than old mice. Pharmacological correction of altered generation of Th17 cell cytokines in immunosenescence represents a novel therapeutic approach to aging-induced inflammatory diseases.
Collapse
Affiliation(s)
- Mei-Chuan Huang
- Department of Medicine, University of California, San Francisco, California, USA
| | | | | | | | | |
Collapse
|
44
|
Thompson SR, Novick D, Stock CJ, Sanders J, Brull D, Cooper J, Woo P, Miller G, Rubinstein M, Humphries SE. Free Interleukin (IL)-18 levels, and the impact of IL18 and IL18BP genetic variation, in CHD patients and healthy men. Arterioscler Thromb Vasc Biol 2007; 27:2743-9. [PMID: 17951325 DOI: 10.1161/atvbaha.107.149245] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate free interleukin-18 (fIL-18) levels, and variation within the IL-18 system genes, in heart surgery patients, and healthy men. METHODS AND RESULTS fIL-18 was calculated from IL-18 and IL-18 binding protein (BP) levels, in 421 healthy men and 196 post-coronary artery bypass graft (CABG) patients. After surgery, fIL-18 peaked at 6 hours (from 117 to 331 pg/mL) but fell to below presurgery levels at 24 hours (99 pg/mL), because of changes in total IL-18 and IL-18BP. fIL-18 24 hours postsurgery was significantly higher in those who suffered a major complication after surgery (125 versus 80 pg/mL, P<0.01). Baseline total IL-18 was also higher in healthy men who went on to suffer an MI over 17 years of prospective study (276 versus 240 pg/mL, P=0.01). Tagging SNPs for IL18 (n=5) and IL18BP (n=3) were determined, in both studies the IL18 HapIII haplotype (frequency 30%) was associated with 36% lower baseline fIL-18 levels before surgery (P<0.01), and 7% lower in healthy men (P=0.04). The frequency of HapIII was lower in CABG patients than in healthy men (20.7 versus 29.8%, P<0.01). CONCLUSIONS IL-18 levels, which are determined in part by variation in IL18, play a role in CHD development and postsurgery outcome.
Collapse
Affiliation(s)
- Simon R Thompson
- Cardiovascular Genetics, Rayne Institute, University College London, WC1E 6JF, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Thompson SR, McCaskie PA, Beilby JP, Hung J, Jennens M, Chapman C, Thompson P, Humphries SE. IL18 haplotypes are associated with serum IL-18 concentrations in a population-based study and a cohort of individuals with premature coronary heart disease. Clin Chem 2007; 53:2078-85. [PMID: 17962365 DOI: 10.1373/clinchem.2007.092692] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Interleukin (IL)-18 is a proinflammatory cytokine that has been implicated in several diseases, including atherosclerosis, and increased circulating IL-18 concentrations increase risk of future coronary heart disease (CHD). We evaluated the effect of common variation within the IL18 gene on concentrations of circulating IL-18. METHODS We measured IL-18, by ELISA, in the population-based study group [Carotid Ultrasound Disease Assessment Study (CUDAS)] and a predominantly male cohort with premature cardiovascular disease [Carotid Ultrasound in Patients with Ischaemic Heart Disease (CUPID)]. Using a tagging single-nucleotide polymorphism (SNP) approach that captured >90% of genetic variation, we identified 4 common (>10%) haplotypes. RESULTS A common SNP was associated with differences in IL-18 concentrations; in CUDAS individuals carrying 2 copies of the rare allele, concentrations were 13% higher than in those with no copies (P = 0.002). Haplotypes were also associated with significant differences in IL-18 concentrations in CUDAS and CUPID. Haplotype GTATA (frequency 23%) was associated with significantly lower IL-18 than others. In CUDAS, those carrying 2 copies had IL-18 concentrations 15% lower than those carrying no copies (P = 0.002); in CUPID, the difference was 22% (P = 0.004). These associations remained significant after adjustment for age, sex, hypertension, HDL cholesterol, waist-to-hip ratio, and alcohol consumption. Despite being associated with differences in IL-18 concentrations, the haplotypes did not occur at different frequencies in those with or without carotid atherosclerotic plaques. CONCLUSIONS Variation within IL18 affects IL-18 concentrations in healthy and diseased individuals and thus may influence the pathophysiology of plaques at all stages of CHD progression.
Collapse
Affiliation(s)
- Simon R Thompson
- Department of Cardiovascular Genetics, University College London, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
In 1988 the task of identifying Type 2 diabetes genes was described as a nightmare. For the next 17 years this proved to be largely correct. In the meantime the prevalence of Type 2 diabetes rose sharply due to non-genetic factors, compounding the problem of trying to find genes. Despite a huge amount of effort, progress was disappointing and only two genes, PPARG and KCNJ11, were confirmed beyond doubt as Type 2 diabetes risk factors in multiple studies. The reasons for this have been well documented and mainly consist of the use of inappropriate levels of statistical inference given the many hundreds of thousands of potential risk polymorphisms in the genome and their small effect sizes. The good news is that these problems are now surmountable and prospects for finding many more genes are bright. This year saw the identification of a third gene, TCF7L2, that has a greater impact on risk than the first two and provided important lessons for Type 2 diabetes genetic studies. The most important of these lessons was that previously unsuspected genes may be involved. In this review I discuss why this year is the start of a new era in our understanding of Type 2 diabetes genes and how this may lead to improved patient care.
Collapse
Affiliation(s)
- T M Frayling
- Peninsula Medical School, University of Exeter, Exeter, UK.
| |
Collapse
|
47
|
Melzer D, Frayling TM, Murray A, Hurst AJ, Harries LW, Song H, Khaw K, Luben R, Surtees PG, Bandinelli SS, Corsi AM, Ferrucci L, Guralnik JM, Wallace RB, Hattersley AT, Pharoah PD. A common variant of the p16(INK4a) genetic region is associated with physical function in older people. Mech Ageing Dev 2007; 128:370-7. [PMID: 17459456 PMCID: PMC2074888 DOI: 10.1016/j.mad.2007.03.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Revised: 03/16/2007] [Accepted: 03/22/2007] [Indexed: 10/23/2022]
Abstract
p16(INK4a) is active in cell senescence, ageing and tumor suppression. Deletion of the small p16(INK4a)/ARF/p15(INK4b) region occurs in many cancers. We screened 25 common polymorphisms across the region and three related genes for associations with physical functioning in older people. In an initial sample of 938 (aged 65-80 years) from the EPIC study (Norfolk, UK), the rs2811712 SNP minor allele (located between the shared p16(INK4a)/ARF locus and p15(INK4b)) was associated with reduced physical impairment. This association remained after testing an additional 1319 EPIC-Norfolk samples (p-value=0.013, total n=2257), and on independent replication in the InCHIANTI study (n=709, p=0.015), and at one-sided significance in Iowa-EPESE (n=419, p=0.079). Overall (n=3372), the prevalence of severely limited physical function was 15.0% in common homozygotes and 7.0% in rare homozygotes (per minor allele odds ratio=1.48, 95% CI: 1.17-1.88, p=0.001, adjusted for age, sex and study). This estimate was similar excluding screening set 1 (OR=1.45, 95% CI: 1.09-1.92, p=0.010, n=2434). These findings require further replication, but provide the first direct evidence that the p16(INK4a)/ARF/p15(INK4b) genetic region and the senescence machinery are active in physical ageing in heterogeneous human populations. The mechanism involved may be via greater cellular restorative activity and reduced stem cell senescence.
Collapse
Affiliation(s)
- David Melzer
- Peninsula Medical School, RD&E Wonford Site, Barrack Road, Exeter EX2 5DW, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|