1
|
Srirangan P, Shyam M, Radhakrishnan V, Prince SE. NLRP3 as a therapeutic target in cyclophosphamide-associated toxicities. Mol Biol Rep 2025; 52:364. [PMID: 40192868 DOI: 10.1007/s11033-025-10479-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/27/2025] [Indexed: 04/20/2025]
Abstract
Cyclophosphamide (CPM), a potent chemotherapeutic agent, while effective against various cancers, can cause significant organ damage. The NLRP3 inflammasome, a key player in the innate immune response, is implicated in this toxicity. This review delves into the intricate relationship between CPM and NLRP3 inflammasome activation, focusing on oxidative stress-mediated organ damage. We explore the mechanisms by which CPM induces NLRP3 activation in the kidneys, heart, liver, and gastrointestinal tract. Additionally, we examine the signaling pathways involved in this process. The review also discusses potential therapeutic interventions, including phytotherapeutic agents, that target NLRP3 inflammasome activation to mitigate CPM-induced organ injury. By highlighting the crucial role of NLRP3 in CPM-related toxicity, this review provides a foundation for future research aimed at developing novel therapeutic strategies to minimize adverse effects and improve patient outcomes.
Collapse
Affiliation(s)
- Prathap Srirangan
- School of Bio Sciences and Technology, VIT University, Vellore, Tamil Nadu, India
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Mukul Shyam
- School of Bio Sciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Vidya Radhakrishnan
- VIT School of Agricultural Innovations and Advanced Learning, VIT University, Vellore, Tamil Nadu, India
| | - Sabina Evan Prince
- School of Bio Sciences and Technology, VIT University, Vellore, Tamil Nadu, India.
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
2
|
Nițulescu IM, Ciulei G, Cozma A, Procopciuc LM, Orășan OH. From Innate Immunity to Metabolic Disorder: A Review of the NLRP3 Inflammasome in Diabetes Mellitus. J Clin Med 2023; 12:6022. [PMID: 37762961 PMCID: PMC10531881 DOI: 10.3390/jcm12186022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
The role of the NLRP3 inflammasome is pivotal in the pathophysiology and progression of diabetes mellitus (DM), encompassing both type 1 (T1D), or type 2 (T2D). As part of the innate immune system, NLRP3 is also responsible for the chronic inflammation triggered by hyperglycemia. In both conditions, NLRP3 facilitates the release of interleukin-1β and interleukin-18. For T1D, NLRP3 perpetuates the autoimmune cascade, leading to the destruction of pancreatic islet cells. In T2D, its activation is associated with the presence of insulin resistance. NLRP3 activation is also instrumental for the presence of numerous complications associated with DM, microvascular and macrovascular. A considerable number of anti-diabetic drugs have demonstrated the ability to inhibit the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Iris Maria Nițulescu
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| | - George Ciulei
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| | - Angela Cozma
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| | - Lucia Maria Procopciuc
- Department 2 of Molecular Sciences, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Olga Hilda Orășan
- Department 4 of Internal Medicine, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.M.N.); (A.C.); (O.H.O.)
| |
Collapse
|
3
|
Ferrara E, Converti I, Scarola R, Tartaglia FC, Gnoni A, Isola G, Rapone B. Mechanism behind the Upregulation of Proteins Associated with the NLRP3 Inflammasome in Periodontitis and Their Role in the Immune Response in Diabetes—A Systematic Review. APPLIED SCIENCES 2023; 13:8278. [DOI: 10.3390/app13148278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background: The molecular crosstalk between periodontitis and diabetes is well established. The role of the NLRP3 inflammasome, a multicomponent inflammatory machinery, is an emerging field of research on the relationship between these two uncommunicable diseases. Recent advances are revealing further molecular details regarding the biological function and the mechanism behind the NLRP3 inflammasome dysregulation and highlighting an unexpected role for the caspase-1 in immune homeostasis. We aimed to understand which metabolic checkpoints are involved in contributing to and instigating the relationship between periodontitis and diabetes. We tried to explore the involvement of the NLRP3 in regulating the cytokine-chemokines profile and discussed the potential synergism in these mechanisms when these two diseases coexist in the same patient. Methods: A literature search was carried out in the electronic databases (MEDLINE, EMBASE, and Cochrane Library) for relevant studies from inception until January 2022 for trials and cohort studies that investigated the activation and regulation mechanism of the NLRP3 inflammasome in patients with periodontitis and type two diabetes. Two investigators independently extracted data. The data quality assessment was rated by the Joanna Briggs Institute (JBI). Results: from twenty-six references identified, three studies (two case-control and one cross-sectional) met the inclusion criteria. Analysis of periodontal tissue samples in diabetic individuals exhibited significant overexpression of the NLRP3 inflammasome when compared with healthy controls. Conclusions: there is insufficient evidence to sustain the involvement of the upregulation of genes and proteins involved in the activation of NLRP3 inflammasome components in patients with periodontitis and diabetes.
Collapse
Affiliation(s)
- Elisabetta Ferrara
- Department of Medical, Oral and Biotechnological Sciences, University G. d’Annunzio, 66100 Chieti, Italy
| | | | | | - Francesco Carlo Tartaglia
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy
| | - Antonio Gnoni
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, “Aldo Moro” University of Bari, 70121 Bari, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Biagio Rapone
- Interdisciplinary Department of Medicine, University of Bari, 70121 Bari, Italy
| |
Collapse
|
4
|
Prevalence of prediabetes, diabetes, diabetes awareness, treatment, and its socioeconomic inequality in west of Iran. Sci Rep 2022; 12:17892. [PMID: 36284227 PMCID: PMC9596718 DOI: 10.1038/s41598-022-22779-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/19/2022] [Indexed: 01/20/2023] Open
Abstract
We aim to estimate the prevalence of prediabetes, and diabetes mellitus (DM). We estimated awareness, treatment, plasma glucose control, and associated factors in diabetes, as well as, socioeconomic-related inequality in the prevalence of diabetes and prediabetes. Data for adults aged 35-70 years were obtained from the baseline phase of the Dehgolan prospective cohort study (DehPCS). Diabetes status was determined as fasting plasma glucose (FPG) of ≥ 126 mg/dl and/or taking glucose lowering medication confirmed by a medical practitioner. Prediabetes was considered as 100 ≤ FPG ≤ 125 mg/dl. The relative concentration index (RCI) was used to exhibit socioeconomic inequality in the prevalence of prediabetes and DM. Prevalence of prediabetes and DM, diabetes awareness and treatment, and glycemic control of DM 18.22%, 10.00%, 78.50%, 68.91% and, 28.50%, respectively. Increasing age (p < 0.001), Increasing body mass index (BMI) (p < 0.05), ex-smoker (p < 0.01), family history of diabetes (FHD) (p < 0.001), and comorbidity (p < 0.001) were independent risk factors for DM. Age group of 46-60 (p < 0.05), ex-smoker (p < 0.05), FHD (p < 0.05) were increased chance of awareness. Current smokers (p < 0.05), and higher education increase the chance of glycemic control in DM. Both DM (RCI = - 0.234) and prediabetes (RCI = - 0.122) were concentrated significantly among less-educated participants. DM was concentrated significantly among poor (RCI = - 0.094) people. A significant proportion of DM awareness and treatment can be due to the integration of diabetes into the primary health care system. The high prevalence of prediabetes and diabetes, which is affected by socioeconomic inequality and combined with low levels of glycemic control may place a greater burden on the health system. Therefore, awareness, receiving treatment, and glycemic control in people with diabetes, and the socioeconomic status of people have become increasingly important in the near future.
Collapse
|
5
|
Hsueh YM, Chen WJ, Chung CJ, Hsieh RL, Chen HH, Huang YL, Shiue HS, Lin MI, Mu SC, Lin YC. The combined effects of nucleotide-binding domain-like receptor protein 3 polymorphisms and levels of blood lead on developmental delays in preschool children. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127317. [PMID: 34879550 DOI: 10.1016/j.jhazmat.2021.127317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/22/2021] [Accepted: 09/19/2021] [Indexed: 06/13/2023]
Abstract
Nucleotide-binding domain-like receptors protein 3 (NLRP3) inflammasomes are associated with neuroinflammation and multiple NLRP3 genes regulate NLRP3 expression. Our study aimed to investigate the association of NLRP3 polymorphisms with developmental delay in preschool children. We also explored whether NLRP3 polymorphisms modified the effects of total urinary arsenic and blood cadmium and lead to developmental delays. A total of 178 children with developmental delays and 88 healthy children were analyzed for urinary arsenic concentrations and red blood cell lead and cadmium concentrations. We examined the genotypes of fifteen common single-nucleotide polymorphisms in NLRP3. We observed that levels of total urinary arsenic and blood lead were significantly associated with developmental delay. The NLRP3rs10754555 CG versus CC/GG, NLRP3rs12048215 AG versus AA/GG, and NLRP3rs12137901 TC/TT versus CC genotype showed a lower odds of developmental delay, with the odds ratio (OR) and 95% confidence interval (CI) = 0.38 (0.19-0.75), 0.52 (0.27-0.99), and 0.33 (0.12-0.90), respectively. Children with the NLRP3rs10754555 CC/GG genotype and high blood lead levels had a significant multiplicative interaction with developmental delay [OR (95% CI) = 9.74 (3.59-26.45)]. This study found evidence that suggested the joint effects of NLRP3rs10754555 CC/GG genotype and high blood lead levels on developmental delays.
Collapse
Affiliation(s)
- Yu-Mei Hsueh
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jen Chen
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Chi-Jung Chung
- Department of Health Risk Management, College of Public Health, China Medical University, Taichung, Taiwan; Department of Medical Research, China Medical University and Hospital, Taichung, Taiwan
| | - Ru-Lan Hsieh
- Department of Physical Medicine and Rehabilitation, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsi-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ya-Li Huang
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Horng-Sheng Shiue
- Department of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-I Lin
- Department of Pediatrics, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Shu-Chi Mu
- Department of Pediatrics, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ying-Chin Lin
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Geriatric Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
6
|
Barbu E, Popescu MR, Popescu AC, Balanescu SM. Inflammation as A Precursor of Atherothrombosis, Diabetes and Early Vascular Aging. Int J Mol Sci 2022; 23:963. [PMID: 35055149 PMCID: PMC8778078 DOI: 10.3390/ijms23020963] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Vascular disease was for a long time considered a disease of the old age, but it is becoming increasingly clear that a cumulus of factors can cause early vascular aging (EVA). Inflammation plays a key role in vascular stiffening and also in other pathologies that induce vascular damage. There is a known and confirmed connection between inflammation and atherosclerosis. However, it has taken a long time to prove the beneficial effects of anti-inflammatory drugs on cardiovascular events. Diabetes can be both a product of inflammation and a cofactor implicated in the progression of vascular disease. When diabetes and inflammation are accompanied by obesity, this ominous trifecta leads to an increased incidence of atherothrombotic events. Research into earlier stages of vascular disease, and documentation of vulnerability to premature vascular disease, might be the key to success in preventing clinical events. Modulation of inflammation, combined with strict control of classical cardiovascular risk factors, seems to be the winning recipe. Identification of population subsets with a successful vascular aging (supernormal vascular aging-SUPERNOVA) pattern could also bring forth novel therapeutic interventions.
Collapse
Affiliation(s)
| | - Mihaela-Roxana Popescu
- Department of Cardiology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 011461 Bucharest, Romania; (E.B.); (S.-M.B.)
| | - Andreea-Catarina Popescu
- Department of Cardiology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 011461 Bucharest, Romania; (E.B.); (S.-M.B.)
| | | |
Collapse
|
7
|
Isola G, Polizzi A, Santonocito S, Alibrandi A, Williams RC. Periodontitis activates the NLRP3 inflammasome in serum and saliva. J Periodontol 2022; 93:135-145. [PMID: 34008185 DOI: 10.1002/jper.21-0049] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Nod-like receptor family pyrin domain-containing protein-3 (NLRP3) complex inflammasome has potentially been shown to play an important role in the development of periodontitis and diabetes. The objective of this study was to analyze the association between serum and salivary NLRP3 concentrations in patients with periodontitis and type-II diabetes mellitus (DM) and to evaluate whether this association was influenced by potential confounders. METHODS For the present study, a cohort of healthy controls (n = 32), and patients with periodontitis (n = 34), type-II DM (n = 33), and a combination of periodontitis + type-II DM (n = 34) were enrolled. Patients were characterized on the basis of their periodontal status and analyzed for demographic characteristics, serum mediators, and for serum and salivary concentrations of NLRP3. A uni- and multivariate model was established to analyze whether periodontitis, type-II DM, and CRP influenced serum and salivary NLRP3 concentrations. RESULTS In comparison to type-II DM patients and healthy controls, patients with periodontitis (serum, P = 0.003; saliva P = 0.012) and periodontitis + type-II DM (serum, P = 0.028; saliva, P = 0.003) had elevated serum and salivary NLRP3 concentrations. The multivariate regression model showed that periodontitis (P = 0.029) and HDL-cholesterol (P = 0.012) were significant predictors of serum NLRP3 concentrations whereas periodontitis (P = 0.036) and CRP (P = 0.012) were significant predictors of salivary NLRP3. CONCLUSION The results of the present study showed that periodontitis and periodontitis + type-II DM patients had higher serum and salivary NLRP3 concentrations in comparison to healthy controls and patients with type-II DM. Periodontitis was demonstrated to be a significant predictor of both serum and salivary NLRP3 concentrations.
Collapse
Affiliation(s)
- Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Surgery and Periodontology, School of Dentistry, University of Catania, Catania, Italy
| | - Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Surgery and Periodontology, School of Dentistry, University of Catania, Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, Unit of Oral Surgery and Periodontology, School of Dentistry, University of Catania, Catania, Italy
| | - Angela Alibrandi
- Department of Economics, Unit of Statistical and Mathematical Sciences, University of Messina, Messina, Italy
| | - Ray C Williams
- Department of Periodontology, UNC-Chapel Hill School of Dentistry, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Özenver N, Efferth T. Phytochemical inhibitors of the NLRP3 inflammasome for the treatment of inflammatory diseases. Pharmacol Res 2021; 170:105710. [PMID: 34089866 DOI: 10.1016/j.phrs.2021.105710] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/15/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023]
Abstract
The NLRP3 inflammasome holds a crucial role in innate immune responses. Pathogen- and danger-associated molecular patterns may initiate inflammasome activation and following inflammatory cytokine release. The inflammasome formation and its-associated activity are involved in various pathological conditions such as cardiovascular, central nervous system, metabolic, renal, inflammatory and autoimmune diseases. Although the mechanism behind NLRP3-mediated disorders have not been entirely illuminated, many phytochemicals and medicinal plants have been described to prevent inflammatory disorders. In the present review, we mainly introduced phytochemicals inhibiting NLRP3 inflammasome in addition to NLRP3-mediated diseases. For this purpose, we performed a systematic literature search by screening PubMed, Scopus, and Google Scholar databases. By compiling the data of phytochemical inhibitors targeting NLRP3 inflammasome activation, a complex balance between inflammasome activation or inhibition with NLRP3 as central player was pointed out in NLRP3-driven pathological conditions. Phytochemicals represent potential therapeutic leads, enabling the generation of chemical derivatives with improved pharmacological features to treat NLRP3-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Nadire Özenver
- Department of Pharmacognosy, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey; Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
9
|
Association of nod-like receptor pyrin domain containing 3 (rs10754558) and protein kinase C zeta (rs2503706) gene polymorphisms with the risk of type 2 diabetes mellitus in Indian population. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
10
|
Pellegrini C, Martelli A, Antonioli L, Fornai M, Blandizzi C, Calderone V. NLRP3 inflammasome in cardiovascular diseases: Pathophysiological and pharmacological implications. Med Res Rev 2021; 41:1890-1926. [PMID: 33460162 DOI: 10.1002/med.21781] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/30/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
Growing evidence points out the importance of nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome in the pathogenesis of cardiovascular diseases (CVDs), including hypertension, myocardial infarct (MI), ischemia, cardiomyopathies (CMs), heart failure (HF), and atherosclerosis. In this regard, intensive research efforts both in humans and in animal models of CVDs are being focused on the characterization of the pathophysiological role of NLRP3 inflammasome signaling in CVDs. In addition, clinical and preclinical evidence is coming to light that the pharmacological blockade of NLRP3 pathways with drugs, including novel chemical entities as well as drugs currently employed in the clinical practice, biologics and phytochemicals, could represent a suitable therapeutic approach for prevention and management of CVDs. On these bases, the present review article provides a comprehensive overview of clinical and preclinical studies about the role of NLRP3 inflammasome in the pathophysiology of CVDs, including hypertension, MI, ischemic injury, CMs, HF and atherosclerosis. In addition, particular attention has been focused on current evidence on the effects of drugs, biologics, and phytochemicals, targeting different steps of inflammasome signaling, in CVDs.
Collapse
Affiliation(s)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | | |
Collapse
|
11
|
CARD8 and IL1B Polymorphisms Influence MRI Brain Patterns in Newborns with Hypoxic-Ischemic Encephalopathy Treated with Hypothermia. Antioxidants (Basel) 2021; 10:antiox10010096. [PMID: 33445495 PMCID: PMC7826682 DOI: 10.3390/antiox10010096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation and oxidative stress are recognized as important contributors of brain injury in newborns due to a perinatal hypoxic-ischemic (HI) insult. Genetic variability in these pathways could influence the response to HI and the outcome of brain injury. The aim of our study was to evaluate the impact of common single-nucleotide polymorphisms in the genes involved in inflammation and response to oxidative stress on brain injury in newborns after perinatal HI insult based on the severity and pattern of magnetic resonance imaging (MRI) findings. The DNA of 44 subjects was isolated from buccal swabs. Genotyping was performed for NLRP3 rs35829419, CARD8 rs2043211, IL1B rs16944, IL1B rs1143623, IL1B rs1071676, TNF rs1800629, CAT rs1001179, SOD2 rs4880, and GPX1 rs1050450. Polymorphism in CARD8 was found to be protective against HI brain injury detected by MRI overall findings. Polymorphisms in IL1B were associated with posterior limb of internal capsule, basal ganglia, and white matter brain patterns determined by MRI. Our results suggest a possible association between genetic variability in inflammation- and antioxidant-related pathways and the severity of brain injury after HI insult in newborns.
Collapse
|
12
|
Cristina de Brito Toscano E, Leandro Marciano Vieira É, Boni Rocha Dias B, Vidigal Caliari M, Paula Gonçalves A, Varela Giannetti A, Maurício Siqueira J, Kimie Suemoto C, Elaine Paraizo Leite R, Nitrini R, Alvarenga Rachid M, Lúcio Teixeira A. NLRP3 and NLRP1 inflammasomes are up-regulated in patients with mesial temporal lobe epilepsy and may contribute to overexpression of caspase-1 and IL-β in sclerotic hippocampi. Brain Res 2020; 1752:147230. [PMID: 33385378 DOI: 10.1016/j.brainres.2020.147230] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/28/2022]
Abstract
Inflammation plays a role in the pathophysiology of mesial temporal lobe epilepsy (MTLE). Inflammasome pathways, including the NLRP1 and NLRP3-induced ones, promote neuroinflammation and pyroptosis through interleukin (IL)-1β and caspase-1 action. Evaluation of NLRP1 in sclerotic hippocampi is scarce and there are no data on NLRP3 in human TLE. The aim of this study was to evaluate the expression of these proteins alongside caspase-1 and IL-1β in the hippocampi of patients with TLE compared to control samples. We also sought to investigate peripheral levels of caspase-1 and IL-1β in an independent cohort. Sclerotic and control hippocampi were collected for both histological and immunohistochemical analyses of NLRP1, NLRP3, caspase-1 and IL-1β; plasma was sampled for the measurement of caspase-1 and IL-1β levels through enzyme-linked immunoassay (ELISA) and cytometric bead array (CBA). Sclerotic hippocampi displayed higher expression of the measured proteins than control. Both glia and neurons showed activation of these pathways. Additionally, increased expression of NLRP1 and NLRP3 was associated with elevated plasma levels of IL-1β and in TLE, and increased levels of peripheral caspase-1 were associated with bilateral hippocampal sclerosis (HS). In conclusion, NLRP1 and NLRP3 are up-regulated in sclerotic hippocampi, what may be responsible, at least in part, for the increased hippocampal expression of caspase-1 and IL-1β. Our data suggest a role for inflammasome activation in central and peripheral inflammation in TLE.
Collapse
Affiliation(s)
- Eliana Cristina de Brito Toscano
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Laboratório Interdisciplinar de Investigação Médica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Érica Leandro Marciano Vieira
- Laboratório Interdisciplinar de Investigação Médica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bárbara Boni Rocha Dias
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcelo Vidigal Caliari
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Paula Gonçalves
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Núcleo Avançado de Tratamento das Epilepsias - Hospital Felício Rocho, Belo Horizonte, MG, Brazil
| | | | - José Maurício Siqueira
- Núcleo Avançado de Tratamento das Epilepsias - Hospital Felício Rocho, Belo Horizonte, MG, Brazil
| | | | | | - Ricardo Nitrini
- Biobank for Aging Studies, Universidade de São Paulo, São Paulo, Brazil
| | - Milene Alvarenga Rachid
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, TX, United States
| |
Collapse
|
13
|
Chen H, Zhang X, Liao N, Ji Y, Mi L, Gan Y, Su Y, Wen F. Identification of NLRP3 Inflammation-Related Gene Promoter Hypomethylation in Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2020; 61:12. [PMID: 33156339 PMCID: PMC7671867 DOI: 10.1167/iovs.61.13.12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose To identify and validate key genes that could provide a new perspective for genetic marker screening of diabetic retinopathy (DR). Methods The gene expression and DNA methylation profiles were obtained from the Gene Expression Omnibus. Differential expression analysis was conducted using the limma package, and then the functions of the differentially expressed genes (DEGs) were analyzed using the DAVID database, followed by protein–protein interaction (PPI) networks using Cytoscape software. We employed the Sequenom MassARRAY system to detect the promoter methylation levels of the candidate genes in peripheral blood mononuclear cells from 32 healthy individuals and 94 patients with type 2 diabetes mellitus (T2D; 64 with DR and 30 without DR) and in fibrovascular membranes (FVMs) from three proliferative DR patients and three controls with idiopathic epiretinal membranes. The mRNA levels of candidate genes were further confirmed via real-time polymerase chain reaction. Results A significant enrichment of 5906 DEGs was found in immune and inflammatory responses. TGFB1, CCL2, and TNFSF2 were identified as the top three core genes associated with NLRP3 inflammation in PPI networks. These genes have relatively low levels of promoter methylation, which have been validated in peripheral blood mononuclear cells and FVMs from DR patients, and the methylation levels were found to be negative correlated with the mRNA levels and HbA1c levels in T2D patients. Conclusions Overall, these data indicate that promoter hypomethylation of NLRP3, TGFB1, CCL2, and TNFSF2 may increase the risk of DR in the Chinese Han population, indicating that these genes might serve as potential targets for the detection and treatment of DR.
Collapse
Affiliation(s)
- Hui Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiongze Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Nanying Liao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuying Ji
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lan Mi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuhong Gan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yongyue Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Feng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Tsetsos F, Roumeliotis A, Tsekmekidou X, Alexouda S, Roumeliotis S, Theodoridis M, Thodis E, Panagoutsos S, Papanas N, Papazoglou D, Kotsa K, Yovos JG, Maltezos E, Passadakis P, Paschou P, Georgitsi M. Genetic variation in CARD8, a gene coding for an NLRP3 inflammasome-associated protein, alters the genetic risk for diabetic nephropathy in the context of type 2 diabetes mellitus. Diab Vasc Dis Res 2020; 17:1479164120970892. [PMID: 33164551 PMCID: PMC7919199 DOI: 10.1177/1479164120970892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Approximately one third of type 2 diabetes mellitus (T2DM) cases present with diabetic nephropathy (DN), the leading cause of end-stage renal disease. Inflammation plays an important role in T2DM disease and DN pathogenesis. NLRP3 inflammasomes are complexes that regulate interleukin-1B (IL-1B) and IL-18 secretion, both involved in inflammatory responses. Activation of NLRP3 is associated with DN onset and progression. Here, we explore whether DN is associated with variants in genes encoding key members of the NLRP3 inflammasome pathway. METHODS Using genome-wide association data, we performed a pilot case-control association study, between 101 DN-T2DM and 185 non-DN-T2DM cases from the Hellenic population across six NLRP3 inflammasome pathway genes. RESULTS Three common CARD8 variants confer decreased risk for DN, namely rs11665831 (OR = 0.62, p = 0.016), rs11083925 (OR = 0.65, p = 0.021), and rs2043211 (OR = 0.66, p = 0.026), independent of sex or co-inheritance with an IL-1B variant. CONCLUSION CARD8 acts as an NLRP3, NF-κB and caspase-1 inhibitor; perhaps, alterations in the cross-talk between CARD8, NF-κB, and NLRP3, which could affect the pro-inflammatory environment in T2DM, render diabetic carriers of certain common CARD8 variants potentially less likely to develop T2DM-related pro-inflammatory responses followed by DN. These preliminary, yet novel, observations will require validation in larger cohorts from several ethnic groups.
Collapse
Affiliation(s)
- Fotis Tsetsos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupoli, Greece
| | - Athanasios Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA University Hospital, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Xanthippi Tsekmekidou
- Division of Endocrinology and Metabolism-Diabetes Center, 1st Department of Internal Medicine, AHEPA University Hospital, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sophia Alexouda
- 1st Laboratory of Medical Biology-Genetics, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA University Hospital, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marios Theodoridis
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Elias Thodis
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Stylianos Panagoutsos
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Nikolaos Papanas
- Diabetes Center, 2nd University Department of Internal Medicine, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Dimitrios Papazoglou
- Diabetes Center, 2nd University Department of Internal Medicine, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism-Diabetes Center, 1st Department of Internal Medicine, AHEPA University Hospital, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - John G Yovos
- Division of Endocrinology and Metabolism-Diabetes Center, 1st Department of Internal Medicine, AHEPA University Hospital, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efstratios Maltezos
- Diabetes Center, 2nd University Department of Internal Medicine, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Ploumis Passadakis
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Peristera Paschou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupoli, Greece
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Marianthi Georgitsi
- 1st Laboratory of Medical Biology-Genetics, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Marianthi Georgitsi, 1st Laboratory of Medical Biology-Genetics, Department of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki 54124, Greece.
| |
Collapse
|
15
|
Herman R, Jensterle M, Janež A, Goričar K, Dolžan V. Genetic Variability in Antioxidative and Inflammatory Pathways Modifies the Risk for PCOS and Influences Metabolic Profile of the Syndrome. Metabolites 2020; 10:metabo10110439. [PMID: 33138337 PMCID: PMC7692942 DOI: 10.3390/metabo10110439] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorder of multifactorial etiopathology likely to involve the interactions between genetics and lifestyle. Chronic inflammation and oxidative stress (OS) may participate in the pathophysiology of the syndrome. The question of the extent to which OS and inflammation are causally related to the development of the syndrome and metabolic complications remains unanswered. By our knowledge, the role of the NLR family pyrin domain containing 3 (NLRP3) inflammasome as an important trigger of inflammatory pathways and NLRP3 and CARD8 polymorphisms has never been addressed in PCOS yet. We conducted a case-control study conducting of total 169 Slovenian PCOS patients and 83 healthy blood donors. They were genotyped for polymorphisms in antioxidative (SOD2 rs4880, CAT rs1001179, PON1 rs854560, and rs662) and inflammatory pathways genes (NLRP3 rs35829419, CARD8 rs2043211, TNF rs1800629, IL1B rs1143623, and rs16944, IL6 rs1800795) using competitive allele-specific polymerase chain reaction (PCR). Logistic regression and the Mann–Whitney test were used in the statistical analysis. SOD2 rs4880, CARD8 rs2043211, and IL1B rs16944 were associated with the risk of developing PCOS. Furthermore, the interactions between CARD8 rs2043211 and IL6 rs1800795 and between IL1B rs1143623 and IL6 rs1800795 also significantly affected the risk for PCOS. With regard to glucose homeostasis, CAT rs1001179, SOD2 rs4880, PON1 rs854560, NLRP3 rs35829419, and TNF rs1800629 were significantly associated with response to the glycemic load. Our data indicate that the genetic variability in the antioxidative and inflammatory pathways influences the development of PCOS and glucose homeostasis in PCOS patients.
Collapse
Affiliation(s)
- Rok Herman
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (R.H.); (M.J.); (A.J.)
| | - Mojca Jensterle
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (R.H.); (M.J.); (A.J.)
- Department of Endocrinology, Diabetes and Metabolic Disease, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Janež
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (R.H.); (M.J.); (A.J.)
- Department of Endocrinology, Diabetes and Metabolic Disease, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
| | - Katja Goričar
- Pharmacogenetics Laboratory, Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Correspondence: ; Tel.: +386-1-543-7670
| |
Collapse
|
16
|
Liu J, Xie X, Yan D, Wang Y, Yuan H, Cai Y, Luo J, Xu A, Huang Y, Cheung CW, Irwin MG, Xia Z. Up-regulation of FoxO1 contributes to adverse vascular remodelling in type 1 diabetic rats. J Cell Mol Med 2020; 24:13727-13738. [PMID: 33108705 PMCID: PMC7754018 DOI: 10.1111/jcmm.15935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Vascular complications from diabetes often result in poor outcomes for patients, even after optimized interventions. Forkhead box protein O1 (FoxO1) is a key regulator of cellular metabolism and plays an important role in vessel formation and maturation. Alterations of FoxO1 occur in the cardiovascular system in diabetes, yet the role of FoxO1 in diabetic vascular complications is poorly understood. In Streptozotocin (STZ)‐induced type 1 diabetic rats, FoxO1 expression was up‐regulated in carotid arteries at 8 weeks of diabetes that was accompanied with adverse vascular remodelling characterized as increased wall thickness, carotid medial cross‐sectional area, media‐to‐lumen ratio and decreased carotid artery lumen area. This adverse vascular remodelling induced by hyperglycaemia in diabetic rats required FoxO1 activation as pharmacological inhibition of FoxO1 with 50mg/kg AS1842856 (AS) reversed vascular remodelling in type 1 diabetic rats. The adverse vascular remodelling in type 1 diabetes mellitus (T1DM) occurred concomitantly with increases in pro‐inflammatory factors, adhesion factors, apoptosis, NOD‐like receptor family protein‐3 inflammasome activation and the phenotypic switch of arterial smooth muscle cells, which were all reversed by AS. In addition, FoxO1 inhibition counteracted the down‐regulation of its upstream mediator PDK1 in T1DM. PDK1 activator reduced FoxO1 nuclear translocation, which serves as the basis for subsequent transcriptional regulation during hyperglycaemia. Taken together, our data suggest that FoxO1 is a critical trigger for type 1 diabetes‐induced vascular remodelling in rats, and inhibition of FoxO1 thus offers a potential therapeutic option for diabetes‐associated cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjin Liu
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Xiang Xie
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China.,Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dan Yan
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Yongshun Wang
- Department of Biomedical Science, University of Hong Kong, Hong Kong, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yin Cai
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Jierong Luo
- Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Heart and Vascular Institute and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Wai Cheung
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Michael G Irwin
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| |
Collapse
|
17
|
Klen J, Goričar K, Dolžan V. Genetic variability in sodium-glucose cotransporter 2 influences glycemic control and risk for diabetic retinopathy in type 2 diabetes patients. J Med Biochem 2020; 39:276-282. [PMID: 33269015 PMCID: PMC7682783 DOI: 10.2478/jomb-2019-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 08/21/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Gluconeogenesis and renal glucose excretion in kidneys both play an important role in glucose homeostasis. Sodium-glucose cotransporter (SGLT2), coded by the SLC5A2 gene is responsible for reabsorption up to 99% of the filtered glucose in proximal tubules. SLC5A2 genetic polymorphisms were suggested to influence glucose homeostasis. We investigated if common SLC5A2 rs9934336 polymorphism influences glycemic control and risk for macro or microvascular complications in Slovenian type 2 diabetes (T2D) patients. METHODS All 181 clinically well characterized T2D patients were genotyped for SLC5A2 rs9934336 G>A polymorphism. Associations with glycemic control and T2D complications were assessed with nonparametric tests and logistic regression. RESULTS SLC5A2 rs9934336 was significantly associated with increased fasting blood glucose levels (P<0.001) and HbA1c levels under the dominant genetic model (P=0.030). After adjustment for T2D duration, significantly higher risk for diabetic retinopathy was present in carriers of at least one polymorphic SLC5A2 rs9934336 A allele compared to non-carriers (OR=7.62; 95%CI=1.65-35.28; P=0.009). CONCLUSIONS Our pilot study suggests an important role of SLC5A2 polymorphisms in the physiologic process of glucose reabsorption in kidneys in T2D patients. This is also the first report on the association between SLC5A2 polymorphism and diabetic retinopathy.
Collapse
Affiliation(s)
- Jasna Klen
- General Hospital Trbovlje, Trbovlje, Slovenia
| | - Katja Goričar
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, Ljubljana, Slovenia
| | - Vita Dolžan
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, Ljubljana, Slovenia
| |
Collapse
|
18
|
Menini S, Iacobini C, Vitale M, Pugliese G. The Inflammasome in Chronic Complications of Diabetes and Related Metabolic Disorders. Cells 2020; 9:E1812. [PMID: 32751658 PMCID: PMC7464565 DOI: 10.3390/cells9081812] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) ranks seventh as a cause of death worldwide. Chronic complications, including cardiovascular, renal, and eye disease, as well as DM-associated non-alcoholic fatty liver disease (NAFLD) account for most of the morbidity and premature mortality in DM. Despite continuous improvements in the management of late complications of DM, significant gaps remain. Therefore, searching for additional strategies to prevent these serious DM-related conditions is of the utmost importance. DM is characterized by a state of low-grade chronic inflammation, which is critical in the progression of complications. Recent clinical trials indicate that targeting the prototypic pro-inflammatory cytokine interleukin-1β (IL-1 β) improves the outcomes of cardiovascular disease, which is the first cause of death in DM patients. Together with IL-18, IL-1β is processed and secreted by the inflammasomes, a class of multiprotein complexes that coordinate inflammatory responses. Several DM-related metabolic factors, including reactive oxygen species, glyco/lipoxidation end products, and cholesterol crystals, have been involved in the pathogenesis of diabetic kidney disease, and diabetic retinopathy, and in the promoting effect of DM on the onset and progression of atherosclerosis and NAFLD. These metabolic factors are also well-established danger signals capable of regulating inflammasome activity. In addition to presenting the current state of knowledge, this review discusses how the mechanistic understanding of inflammasome regulation by metabolic danger signals may hopefully lead to novel therapeutic strategies targeting inflammation for a more effective treatment of diabetic complications.
Collapse
Affiliation(s)
| | | | | | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (S.M.); (C.I.); (M.V.)
| |
Collapse
|
19
|
van der Made CI, Hoischen A, Netea MG, van de Veerdonk FL. Primary immunodeficiencies in cytosolic pattern-recognition receptor pathways: Toward host-directed treatment strategies. Immunol Rev 2020; 297:247-272. [PMID: 32640080 DOI: 10.1111/imr.12898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
In the last decade, the paradigm of primary immunodeficiencies (PIDs) as rare recessive familial diseases that lead to broad, severe, and early-onset immunological defects has shifted toward collectively more common, but sporadic autosomal dominantly inherited isolated defects in the immune response. Patients with PIDs constitute a formidable area of research to study the genetics and the molecular mechanisms of complex immunological pathways. A significant subset of PIDs affect the innate immune response, which is a crucial initial host defense mechanism equipped with pattern-recognition receptors. These receptors recognize pathogen- and damage-associated molecular patterns in both the extracellular and intracellular space. In this review, we will focus on primary immunodeficiencies caused by genetic defects in cytosolic pattern-recognition receptor pathways. We discuss these PIDs organized according to their mutational mechanisms and consequences for the innate host response. The advanced understanding of these pathways obtained by the study of PIDs creates the opportunity for the development of new host-directed treatment strategies.
Collapse
Affiliation(s)
- Caspar I van der Made
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud Institute of Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands.,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander Hoischen
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud Institute of Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands.,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud Institute of Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud Institute of Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
20
|
Baragetti A, Catapano AL, Magni P. Multifactorial Activation of NLRP3 Inflammasome: Relevance for a Precision Approach to Atherosclerotic Cardiovascular Risk and Disease. Int J Mol Sci 2020; 21:E4459. [PMID: 32585928 PMCID: PMC7352274 DOI: 10.3390/ijms21124459] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic low-grade inflammation, through the specific activation of the NACHT leucine-rich repeat- and PYD-containing (NLRP)3 inflammasome-interleukin (IL)-1β pathway, is an important contributor to the development of atherosclerotic cardiovascular disease (ASCVD), being triggered by intracellular cholesterol accumulation within cells. Within this pathological context, this complex pathway is activated by a number of factors, such as unhealthy nutrition, altered gut and oral microbiota, and elevated cholesterol itself. Moreover, evidence from autoinflammatory diseases, like psoriasis and others, which are also associated with higher cardiovascular disease (CVD) risk, suggests that variants of NLRP3 pathway-related genes (like NLRP3 itself, caspase recruitment domain-containing protein (CARD)8, caspase-1 and IL-1β) may carry gain-of-function mutations leading, in some individuals, to a constitutive pro-inflammatory pattern. Indeed, some reports have recently associated the presence of specific single nucleotide polymorphisms (SNPs) on such genes with greater ASCVD prevalence. Based on these observations, a potential effective strategy in this context may be the identification of carriers of these NLRP3-related SNPs, to generate a genomic score, potentially useful for a better CVD risk prediction, and, possibly, for personalized therapeutic approaches targeted to the NLRP3-IL-1β pathway.
Collapse
Affiliation(s)
- Andrea Baragetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (A.B.); (A.L.C.)
- SISA, Center for the Study of Atherosclerosis, Bassini Hospital, 20092 Cinisello Balsamo, Italy
| | - Alberico Luigi Catapano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (A.B.); (A.L.C.)
- IRCCS Multimedica Hospital, 20099 Milan, Italy
| | - Paolo Magni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (A.B.); (A.L.C.)
- IRCCS Multimedica Hospital, 20099 Milan, Italy
| |
Collapse
|
21
|
Effect of Quamoclit angulata Extract Supplementation on Oxidative Stress and Inflammation on Hyperglycemia-Induced Renal Damage in Type 2 Diabetic Mice. Antioxidants (Basel) 2020; 9:antiox9060459. [PMID: 32471242 PMCID: PMC7346142 DOI: 10.3390/antiox9060459] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is caused by abnormalities of controlling blood glucose and insulin homeostasis. Especially, hyperglycemia causes hyper-inflammation through activation of NLRP3 inflammasome, which can lead to cell apoptosis, hypertrophy, and fibrosis. Quamoclit angulata (QA), one of the annual winders, has been shown ameliorative effects on diabetes. The current study investigated whether the QA extract (QAE) attenuated hyperglycemia-induced renal inflammation related to NLRP inflammasome and oxidative stress in high fat diet (HFD)-induced diabetic mice. After T2DM was induced, the mice were treated with QAE (5 or 10 mg/kg/day) by gavage for 12 weeks. The QAE supplementation reduced homeostasis model assessment insulin resistance (HOMA-IR), kidney malfunction, and glomerular hypertrophy in T2DM. Moreover, the QAE treatment significantly attenuated renal NLRP3 inflammasome dependent hyper-inflammation and consequential renal damage caused by oxidative stress, apoptosis, and fibrosis in T2DM. Furthermore, QAE normalized aberrant energy metabolism (downregulation of p-AMPK, sirtuin (SIRT)-1, and PPARγ-coactivator α (PGC-1 α)) in T2DM mice. Taken together, the results suggested that QAE as a natural product has ameliorative effects on renal damage by regulation of oxidative stress and inflammation in T2DM.
Collapse
|
22
|
Lespedeza bicolor Extract Ameliorated Renal Inflammation by Regulation of NLRP3 Inflammasome-Associated Hyperinflammation in Type 2 Diabetic Mice. Antioxidants (Basel) 2020; 9:antiox9020148. [PMID: 32050658 PMCID: PMC7071116 DOI: 10.3390/antiox9020148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by hyperglycemia. The chronic hyperglycemic condition causes hyperinflammation via activation of nucleotide-binding oligomerization domain-like pyrin domain containing receptor 3 (NLRP3) inflammasome and abnormally leads to morphological and functional changes in kidney. A previous study showed a protective effect of Lespedeza bicolor extract (LBE) on endothelial dysfunction induced by methylglyoxal glucotoxicity. We aimed to investigate whether LBE ameliorated renal damage through regulation of NLRP3 inflammasome-dependent hyper-inflammation in T2DM mice. After T2DM induction by a high fat diet and low dose of streptozotocin (30 mg/kg), the mice were administered with different dosages of LBE (100 or 250 mg/kg/day) by gavage for 12 weeks. LBE supplementation ameliorated kidney dysfunction demonstrated by urine albumin-creatinine at a low dose and plasma creatinine, blood urea nitrogen (BUN), and glomerular hypertrophy at a high dose. Furthermore, a high dose of LBE supplementation significantly attenuated renal hyper-inflammation associated with NLRP3 inflammasome and oxidative stress related to nuclear factor erythroid 2-related factor 2 (Nrf-2) in T2DM mice. Meanwhile, a low dose of LBE supplementation up-regulated energy metabolism demonstrated by phosphorylation of adenosine monophosphate kinase (AMPK) and Sirtuin (SIRT)-1 in T2DM mice. In conclusion, the current study suggested that LBE, in particular, at a high dose could be used as a beneficial therapeutic for hyperglycemia-induced renal damage in T2DM.
Collapse
|
23
|
Capozzi ME, Savage SR, McCollum GW, Hammer SS, Ramos CJ, Yang R, Bretz CA, Penn JS. The peroxisome proliferator-activated receptor-β/δ antagonist GSK0660 mitigates retinal cell inflammation and leukostasis. Exp Eye Res 2020; 190:107885. [PMID: 31758977 PMCID: PMC7426872 DOI: 10.1016/j.exer.2019.107885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022]
Abstract
Diabetic retinopathy (DR) is triggered by retinal cell damage stimulated by the diabetic milieu, including increased levels of intraocular free fatty acids. Free fatty acids may serve as an initiator of inflammatory cytokine release from Müller cells, and the resulting cytokines are potent stimulators of retinal endothelial pathology, such as leukostasis, vascular permeability, and basement membrane thickening. Our previous studies have elucidated a role for peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) in promoting several steps in the pathologic cascade in DR, including angiogenesis and expression of inflammatory mediators. Furthermore, PPARβ/δ is a known target of lipid signaling, suggesting a potential role for this transcription factor in fatty acid-induced retinal inflammation. Therefore, we hypothesized that PPARβ/δ stimulates both the induction of inflammatory mediators by Müller cells as well the paracrine induction of leukostasis in endothelial cells (EC) by Müller cell inflammatory products. To test this, we used the PPARβ/δ inhibitor, GSK0660, in primary human Müller cells (HMC), human retinal microvascular endothelial cells (HRMEC) and mouse retina. We found that palmitic acid (PA) activation of PPARβ/δ in HMC leads to the production of pro-angiogenic and/or inflammatory cytokines that may constitute DR-relevant upstream paracrine inflammatory signals to EC and other retinal cells. Downstream, EC transduce these signals and increase their synthesis and release of chemokines such as CCL8 and CXCL10 that regulate leukostasis and other cellular events related to vascular inflammation in DR. Our results indicate that PPARβ/δ inhibition mitigates these upstream (MC) as well as downstream (EC) inflammatory signaling events elicited by metabolic stimuli and inflammatory cytokines. Therefore, our data suggest that PPARβ/δ inhibition is a potential therapeutic strategy against early DR pathology.
Collapse
Affiliation(s)
- Megan E Capozzi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, USA.
| | - Sara R Savage
- Department of Pharmacology, Vanderbilt University, USA
| | - Gary W McCollum
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA
| | - Sandra S Hammer
- Department of Cell and Developmental Biology, Vanderbilt University, USA
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA
| | - Rong Yang
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA
| | - Colin A Bretz
- Department of Cell and Developmental Biology, Vanderbilt University, USA
| | - John S Penn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, USA; Department of Pharmacology, Vanderbilt University, USA; Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA; Department of Cell and Developmental Biology, Vanderbilt University, USA
| |
Collapse
|
24
|
Murphy AM, Smith CE, Murphy LM, Follis JL, Tanaka T, Richardson K, Noordam R, Lemaitre RN, Kähönen M, Dupuis J, Voortman T, Marouli E, Mook‐Kanamori DO, Raitakari OT, Hong J, Dehghan A, Dedoussis G, de Mutsert R, Lehtimäki T, Liu C, Rivadeneira F, Deloukas P, Mikkilä V, Meigs JB, Uitterlinden A, Ikram MA, Franco OH, Hughes M, O' Gaora P, Ordovás JM, Roche HM. Potential Interplay between Dietary Saturated Fats and Genetic Variants of the NLRP3 Inflammasome to Modulate Insulin Resistance and Diabetes Risk: Insights from a Meta-Analysis of 19 005 Individuals. Mol Nutr Food Res 2019; 63:e1900226. [PMID: 31432628 PMCID: PMC6864231 DOI: 10.1002/mnfr.201900226] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/12/2019] [Indexed: 12/13/2022]
Abstract
SCOPE Insulin resistance (IR) and inflammation are hallmarks of type 2 diabetes (T2D). The nod-like receptor pyrin domain containing-3 (NLRP3) inflammasome is a metabolic sensor activated by saturated fatty acids (SFA) initiating IL-1β inflammation and IR. Interactions between SFA intake and NLRP3-related genetic variants may alter T2D risk factors. METHODS Meta-analyses of six Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium (n = 19 005) tested interactions between SFA and NLRP3-related single-nucleotide polymorphisms (SNPs) and modulation of fasting insulin, fasting glucose, and homeostasis model assessment of insulin resistance. RESULTS SFA interacted with rs12143966, wherein each 1% increase in SFA intake increased insulin by 0.0063 IU mL-1 (SE ± 0.002, p = 0.001) per each major (G) allele copy. rs4925663, interacted with SFA (β ± SE = -0.0058 ± 0.002, p = 0.004) to increase insulin by 0.0058 IU mL-1 , per additional copy of the major (C) allele. Both associations are close to the significance threshold (p < 0.0001). rs4925663 causes a missense mutation affecting NLRP3 expression. CONCLUSION Two NLRP3-related SNPs showed potential interaction with SFA to modulate fasting insulin. Greater dietary SFA intake accentuates T2D risk, which, subject to functional validation, may be further elaborated depending on NLRP3-related genetic variants.
Collapse
Affiliation(s)
- Aoife M. Murphy
- Nutrigenomics Research GroupConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - Caren E. Smith
- Jean Mayer USDA Human Nutrition Research Centre on AgingTufts UniversityBostonMA02111USA
| | - Leanne M. Murphy
- UCD School of Biomolecular and Biomedical ScienceConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - Jack L. Follis
- Department of MathematicsUniversity of St. ThomasHoustonTX77006‐4626USA
| | - Toshiko Tanaka
- Translational Gerontology BranchNational Institute on AgingBaltimoreMD21224USA
| | - Kris Richardson
- Jean Mayer USDA Human Nutrition Research Centre on AgingTufts UniversityBostonMA02111USA
| | - Raymond Noordam
- Department of Internal MedicineSection of Gerontology and Geriatrics, Leiden University Medical CenterLeiden2333 ZA.The Netherlands
| | | | - Mika Kähönen
- Department of Clinical PhysiologyTampere University Hospital and University of Tampere School of Medicine33521TampereFinland
| | - Josée Dupuis
- Department of BiostatisticsBoston University School of Public HealthBostonMA02130USA
| | - Trudy Voortman
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Eirini Marouli
- William Harvey Research InstituteBarts and The London School of Medicine and DentistryQueen Mary University of LondonLondonE1 4NSUK
| | - Dennis O. Mook‐Kanamori
- Department of Clinical Epidemiology and Department of Public Health and Primary CareLeiden University Medical CenterAlbinusdreef 22333 ZALeidenThe Netherlands
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear MedicineTurku University Hospital, and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku20521TurkuFinland
| | - Jaeyoung Hong
- Department of BiostatisticsBoston University School of Public HealthBostonMA02130USA
| | - Abbas Dehghan
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - George Dedoussis
- Department of Nutrition and DieteticsSchool of Health Science and Education, Harokopio UniversityEl. Venizelou 7017671AthensGreece
| | - Renée de Mutsert
- Department of Clinical Epidemiology and Department of Public Health and Primary CareLeiden University Medical CenterAlbinusdreef 22333 ZALeidenThe Netherlands
| | - Terho Lehtimäki
- Department of Clinical ChemistryFimlab Laboratories and Finnish Cardiovascular Research Center–TampereFaculty of Medicine and Life Sciences, University of TampereTampere33520Finland
| | - Ching‐Ti Liu
- Department of BiostatisticsBoston University School of Public HealthBostonMA02130USA
| | - Fernando Rivadeneira
- Department of Internal MedicineErasmus University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Panagiotis Deloukas
- William Harvey Research InstituteBarts and The London School of Medicine and DentistryQueen Mary University of LondonLondonE1 4NSUK
| | - Vera Mikkilä
- Division of NutritionDepartment of Food and Environmental Sciences00014HelsinkiFinland
| | - James B. Meigs
- Division of General Internal MedicineMassachusetts General HospitalBostonMA02114USA
- Harvard Medical SchoolBostonMA02115USA
- Broad InstituteCambridgeMA02142USA
| | - Andre Uitterlinden
- Department of Internal MedicineErasmus University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Mohammad A. Ikram
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Oscar H. Franco
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Maria Hughes
- Nutrigenomics Research GroupConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - Peadar O' Gaora
- UCD School of Biomolecular and Biomedical ScienceConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - José M. Ordovás
- Jean Mayer USDA Human Nutrition Research Centre on AgingTufts UniversityBostonMA02111USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)28029MadridSpain
- IMDEA Food Institute, CEI UAM + CSICE ‐ 28049MadridSpain
| | - Helen M. Roche
- Nutrigenomics Research GroupConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
- Institute For Global Food SecurityQueen's University BelfastNorthern Ireland
| |
Collapse
|
25
|
Šoštarič A, Jenko B, Kozjek NR, Ovijač D, Šuput D, Milisav I, Dolžan V. Detection of metabolic syndrome burden in healthy young adults may enable timely introduction of disease prevention. Arch Med Sci 2019; 15:1184-1194. [PMID: 31572463 PMCID: PMC6764305 DOI: 10.5114/aoms.2019.87462] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/03/2018] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Metabolic syndrome and associated diseases are a global health problem. Detection of early metabolic modifications that may lead to metabolic syndrome would enable timely introduction of preventive lifestyle modifications. MATERIAL AND METHODS In total 103 young, healthy adults were assessed for indicators of metabolic alterations. Anthropometric, lifestyle, genetic and biochemical parameters were assessed. Individuals who fulfilled at least one criterion for diagnosis of metabolic syndrome were assigned to the group with the higher metabolic syndrome burden (B-MeS). RESULTS The 34 young healthy individuals who were assigned to the B-MeS group had lower fat-free mass, higher body mass index, waist-to-hip ratio, fat mass, and blood pressure, more visceral fat, they were less physically active, had higher C-reactive protein values and higher catalase activity. Their phenotype was more similar to that of patients diagnosed with metabolic syndrome than the rest of the population. CONCLUSIONS Simple anthropometric measurements, lifestyle assessment and basic biochemical measurements can be used to identify young healthy individuals with increased risk for metabolic syndrome. These assessments can be performed at periodic check-ups of the healthy population so that timely diagnosis of B-MeS can be made. As lifestyle factors have a big influence on development or improvement of the MeS, the timely diagnosis for B-MeS would enable an early opportunity for intervention for lifestyle modification in the still healthy population, saving costs and reducing disability adjusted life years.
Collapse
Affiliation(s)
- Anja Šoštarič
- Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Barbara Jenko
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nada Rotovnik Kozjek
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Institute of Oncology, Ljubljana, Slovenia
| | - Darja Ovijač
- Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
| | - Dušan Šuput
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Irina Milisav
- Faculty of Health Sciences, University of Ljubljana, Ljubljana, Slovenia
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vita Dolžan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
26
|
Yi X, Zhang L, Lu W, Tan X, Yue J, Wang P, Xu W, Ye L, Huang D. The effect of NLRP inflammasome on the regulation of AGEs-induced inflammatory response in human periodontal ligament cells. J Periodontal Res 2019; 54:681-689. [PMID: 31250434 DOI: 10.1111/jre.12677] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/18/2019] [Accepted: 06/01/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Diabetes influences the frequency and development of periodontitis. Inflammation of human periodontal ligament cells (HPDLCs) participates in this pathologic process. Hence, this study aims to explore whether advanced glycation end products (AGEs), by-products of diabetes, could exaggerate inflammation induced by muramyl dipeptide (MDP) in HPDLCs, and whether nucleotide-binding oligomerization domain-like receptors (NLRs) signaling pathway was involved. MATERIAL AND METHODS Human periodontal ligament cells were pre-treated with 100 μg/mL AGEs for 24 hours and stimulated with 10 μg/mL MDP for 24 hours. IL-6, IL-1β, and RAGE were detected, and the activation of NF-κB signaling pathway was observed. The expression of NLRs was evaluated with or without silencing RAGE or blocking NF-κB pathway under AGEs stimulation. Statistical analyses were performed by using independent sample t test. RESULTS Advanced glycation end products induced significant increase of inflammatory cytokines in HPDLCs (P < 0.05). Results of western blot (WB) showed that after 45 minutes stimulation of AGEs, p-p65/p65 ratio peaked; AGEs promoted the expression of NLRP1, NLRP3, and apoptosis-associated speck-like protein containing a CARD (ASC). After silencing RAGE or blocking NF-κB pathway, the up-regulation of NLRs protein caused by AGEs was attenuated. Additionally, AGEs pre-treatment could enhance the inflammatory response of MDP and the expression of NLRs, which were demonstrated by more expression of IL-6, IL-1β, NOD2, NLRP1, NLRP3, and ASC. CONCLUSION Advanced glycation end products induced inflammatory response in HPDLCs via NLRP1-inflammasome and NLRP3-inflammasome activation in which NF-κB signal pathway was involved. Besides, AGEs promoted the inflammatory response of MDP via NOD2, NLRP1-inflammasome, and NLRP3-inflammasome.
Collapse
Affiliation(s)
- Xiaowei Yi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wanlu Lu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junli Yue
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Hospital of Stomatology Wuhan University, Wuhan, China
| | - Puyu Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,The Affiliated Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Weizhe Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Ward R, Li W, Abdul Y, Jackson L, Dong G, Jamil S, Filosa J, Fagan SC, Ergul A. NLRP3 inflammasome inhibition with MCC950 improves diabetes-mediated cognitive impairment and vasoneuronal remodeling after ischemia. Pharmacol Res 2019; 142:237-250. [PMID: 30818045 PMCID: PMC6486792 DOI: 10.1016/j.phrs.2019.01.035] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/07/2019] [Accepted: 01/17/2019] [Indexed: 12/11/2022]
Abstract
Diabetes increases the risk and worsens the progression of cognitive impairment via the greater occurrence of small vessel disease and stroke. Yet, the underlying mechanisms are not fully understood. It is now accepted that cardiovascular health is critical for brain health and any neurorestorative approaches to prevent/delay cognitive deficits should target the conceptual neurovascular unit (NVU) rather than neurons alone. We have recently shown that there is augmented hippocampal NVU remodeling after a remote ischemic injury in diabetes. NLRP3 inflammasome signaling has been implicated in the development of diabetes and neurodegenerative diseases, but little is known about the impact of NLRP3 activation on functional and structural interaction within the NVU of hippocampus, a critical part of the brain that is involved in forming, organizing, and storing memories. Endothelial cells are at the center of the NVU and produce trophic factors such as brain derived neurotrophic factor (BDNF) contributing to neuronal survival, known as vasotrophic coupling. Therefore, the aims of this study focused on two hypotheses: 1) diabetes negatively impacts hippocampal NVU remodeling and worsens cognitive outcome after stroke, and 2) NLRP3 inhibition with MCC950 will improve NVU remodeling and cognitive outcome following stroke via vasotrophic (un)coupling between endothelial cells and hippocampal neurons. Stroke was induced through a 90-min transient middle cerebral artery occlusion (MCAO) in control and high-fat diet/streptozotocin-induced (HFD/STZ) diabetic male Wistar rats. Saline or MCC950 (3 mg/kg), an inhibitor of NLRP3, was injected at 1 and 3 h after reperfusion. Cognition was assessed over time and neuronal density, blood-brain barrier (BBB) permeability as well as NVU remodeling (aquaporin-4 [AQP4] polarity) was measured on day 14 after stroke. BDNF was measured in endothelial and hippocampal neuronal cultures under hypoxic and diabetes-mimicking condition with and without NLRP3 inhibition. Diabetes increased neuronal degeneration and BBB permeability, disrupted AQP4 polarity, impaired cognitive function and amplified NLRP3 activation after ischemia. Inhibition with MCC950 improved cognitive function and vascular integrity after stroke in diabetic animals and prevented hypoxia-mediated decrease in BDNF secretion. These results are the first to provide essential data showing MCC950 has the potential to become a therapeutic to prevent neurovascular remodeling and worsened cognitive decline in diabetic patients following stroke.
Collapse
Affiliation(s)
- Rebecca Ward
- Departments of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Weiguo Li
- Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, United States
| | - Yasir Abdul
- Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, United States
| | - LaDonya Jackson
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Guangkuo Dong
- Physiology, Augusta University, Augusta, GA, United States
| | - Sarah Jamil
- Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jessica Filosa
- Physiology, Augusta University, Augusta, GA, United States
| | - Susan C Fagan
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Adviye Ergul
- Physiology, Augusta University, Augusta, GA, United States; Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
28
|
Wang R, Zhang P, Li Z, Lv X, Cai H, Gao C, Song Y, Yu Y, Li B, Cui Y. The prevalence of pre-diabetes and diabetes and their associated factors in Northeast China: a cross-sectional study. Sci Rep 2019; 9:2513. [PMID: 30792436 PMCID: PMC6385189 DOI: 10.1038/s41598-019-39221-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 12/27/2018] [Indexed: 02/05/2023] Open
Abstract
This study investigates the prevalence of pre-diabetes and diabetes and their associated risk factors among adults in Northeast China. A multistage stratified cluster sampling method was used to select adults from Jilin Province. Out of an initial recruitment of 23,050 individuals, 21,435 participants completed an interview and medical examination. The estimated prevalence of diabetes and pre-diabetes were 9.1% and 19.8%, respectively. The prevalence of hypertension, dyslipidemia, and obesity were the highest in participants with previously diagnosed diabetes. Participants who were previously diagnosed with diabetes were more likely to be aware of their hypertension and dyslipidemia status. Participants who were older, male, more educated, or who were widows or widowers were at greater risk for pre-diabetes. Similarly, those who were current drinkers or smokers, had higher BMI or waist circumference, had a family history of diabetes, or who reported they lived in urban areas or had low physical activity levels had increased pre-diabetes risk. The observed levels of diabetes and pre-diabetes in this study indicate that the medical authority needs to focus more attention in this area, and that health monitoring is essential to improving the health awareness of its residents.
Collapse
Affiliation(s)
- Rui Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Peng Zhang
- Department of Neurology, Stroke Center, the First Hospital of Jilin University, Chang Chun, 130021, China
| | - Zhijun Li
- Department of Epidemiology and Biostatistics, Beihua University School of Public Health, Chang Chun, China
| | - Xin Lv
- Department of Epidemiology and Biostatistics, Jilin University School of Public Health, 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Hongfei Cai
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chunshi Gao
- Department of Epidemiology and Biostatistics, Jilin University School of Public Health, 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Yuanyuan Song
- Department of Epidemiology and Biostatistics, Jilin University School of Public Health, 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Yaqin Yu
- Department of Epidemiology and Biostatistics, Jilin University School of Public Health, 1163 Xinmin Street, Changchun, Jilin, 130021, China
| | - Bo Li
- Department of Epidemiology and Biostatistics, Jilin University School of Public Health, 1163 Xinmin Street, Changchun, Jilin, 130021, China.
| | - Youbin Cui
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
29
|
Trotta MC, Maisto R, Guida F, Boccella S, Luongo L, Balta C, D’Amico G, Herman H, Hermenean A, Bucolo C, D’Amico M. The activation of retinal HCA2 receptors by systemic beta-hydroxybutyrate inhibits diabetic retinal damage through reduction of endoplasmic reticulum stress and the NLRP3 inflammasome. PLoS One 2019; 14:e0211005. [PMID: 30657794 PMCID: PMC6338370 DOI: 10.1371/journal.pone.0211005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE The role of the hydroxycarboxylic acid receptor 2 (HCA2) in the retinal damage induced by diabetes has never been explored. In this context, the present study highlights an upregulation of retinal HCA2 receptors in diabetic C57BL6J mice. Moreover, we illustrate that HCA2 receptors exert an anti-inflammatory effect on the retinal damage induced by diabetes when activated by the endogenous ligand β-hydroxybutyrate. METHODOLOGY Seven-to-10-week-old C57BL6J mice were rendered diabetic by a single intraperitoneal injection of streptozotocin (75 mg/kg of body weight) and monitored intermittently over a 10-week period extending from the initial diabetes assessment. Mice with a fasting blood glucose level higher than 250 mg/dl for 2 consecutive weeks after streptozotocin injection were treated twice a week with intraperitoneal injections of 25-50-100 mg/kg β-hydroxybutyrate. RESULTS Interestingly, while the retinal endoplasmic reticulum stress markers (pPERK, pIRE1, ATF-6α) were elevated in diabetic C57BL6J mice, their levels were significantly reduced by the systemic intraperitoneal treatment with 50 mg/kg and 100 mg/kg β-hydroxybutyrate. These mice also exhibited high NLRP3 inflammasome activity and proinflammatory cytokine levels. In fact, the elevated levels of retinal NLRP3 inflammasome activation markers (NLRP3, ASC, caspase-1) and of the relative proinflammatory cytokines (IL-1β, IL-18) were significantly reduced by 50 mg/kg and 100 mg/kg β-hydroxybutyrate treatment. These doses also reduced the high apoptotic cell number exhibited by the diabetic mice in the retinal outer nuclear layer (ONL) and increased the ONL low connexin 43 expression, leading to an improvement in retinal permeability and homeostasis. CONCLUSIONS These data suggest that the systemic treatment of diabetic C57BL6J mice with BHB activates retinal HCA2 and inhibits local damage.
Collapse
Affiliation(s)
- Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Cornel Balta
- Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | | | - Hildegard Herman
- Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
| | - Anca Hermenean
- Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
- Center for Research in Ocular Pharmacology—CERFO University of Catania, Catania, Italy
| | - Michele D’Amico
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
- * E-mail:
| |
Collapse
|
30
|
Smigoc Schweiger D, Goricar K, Hovnik T, Mendez A, Bratina N, Brecelj J, Vidan-Jeras B, Battelino T, Dolzan V. Dual Role of PTPN22 but Not NLRP3 Inflammasome Polymorphisms in Type 1 Diabetes and Celiac Disease in Children. Front Pediatr 2019; 7:63. [PMID: 30915320 PMCID: PMC6422865 DOI: 10.3389/fped.2019.00063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/18/2019] [Indexed: 12/15/2022] Open
Abstract
Genetic polymorphisms in genes coding for inflammasome components nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain-containing protein 3 (NLRP3) and caspase recruitment domain-containing protein 8 (CARD8) have been associated with autoinflammatory and autoimmune diseases. On the other hand several studies suggested that NLRP3 inflammasome contributes to maintenance of gastrointestinal immune homeostasis and that activation of NLRP3 is regulated by protein tyrosine phosphatase non-receptor 22 (PTPN22). PTPN22 polymorphism was implicated in the risk for various autoimmune diseases including type 1 diabetes (T1D) but not for celiac disease (CD). The aim of our study was to evaluate the role of inflammasome related polymorphisms in subjects with either T1D or CD as well as in subjects affected by both diseases. We examined PTPN22 rs2476601 (p.Arg620Trp), NLRP3 rs35829419 (p.Gln705Lys), and CARD8 rs2043211 (p.Cys10Ter) in 66 subjects with coexisting T1D and CD, 65 subjects with T1D who did not develop CD, 67 subjects diagnosed only with CD and 127 healthy unrelated Slovenian individuals. All results were adjusted for clinical characteristic and human leukocyte antigen (HLA) risk. PTPN22 rs2476601 allele was significantly more frequent among subjects with T1D (Padj = 0.001) and less frequent in subjects with CD (Padj = 0.039) when compared to controls. In patients with coexisting T1D and CD this variant was significantly less frequent compared to T1D group (Padj = 0.010). Protective effect on CD development in individuals with T1D was observed only within the low risk HLA group. On the other hand, we found no association of NLRP3 rs35829419 and CARD8 rs2043211 with the development of T1D, CD or both diseases together. In conclusion PTPN22 rs2476601polymorphism was significantly associated with the risk of developing T1D in Slovenian population, while no associations of proinflammatory NLRP3 and CARD8 polymorphisms with T1D and CD were observed. Interestingly, the same PTPN22 variant protected from CD. We hypothesize that this effect may be mediated through the NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Darja Smigoc Schweiger
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Katja Goricar
- Pharmacogenetics Laboratory, Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Tinka Hovnik
- Unit of Special Laboratory Diagnostics, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Andrijana Mendez
- Tissue Typing Centre, Blood Transfusion Center of Slovenia, Ljubljana, Slovenia
| | - Natasa Bratina
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Brecelj
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Department of Gastroenterology, Hepatology and Nutrition, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Blanka Vidan-Jeras
- Tissue Typing Centre, Blood Transfusion Center of Slovenia, Ljubljana, Slovenia
| | - Tadej Battelino
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vita Dolzan
- Pharmacogenetics Laboratory, Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
31
|
Imani D, Azimi A, Salehi Z, Rezaei N, Emamnejad R, Sadr M, Izad M. Association of nod-like receptor protein-3 single nucleotide gene polymorphisms and expression with the susceptibility to relapsing-remitting multiple sclerosis. Int J Immunogenet 2018; 45:329-336. [DOI: 10.1111/iji.12401] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 05/13/2018] [Accepted: 08/16/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Danyal Imani
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Amirreza Azimi
- Neurology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
- MS Research Center, Neuroscience Institute; Tehran University of Medical Sciences; Tehran Iran
| | - Zahra Salehi
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Nima Rezaei
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
- Molecular Immunology Research Center; Tehran University of Medical Sciences; Tehran Iran
| | - Rahimeh Emamnejad
- Immunology Department, School of Medicine; Shahre-Kord University of Medical Sciences, Shahre-Kord; Iran
| | - Maryam Sadr
- Molecular Immunology Research Center; Tehran University of Medical Sciences; Tehran Iran
| | - Maryam Izad
- Immunology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
- MS Research Center, Neuroscience Institute; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
32
|
Rovira-Llopis S, Apostolova N, Bañuls C, Muntané J, Rocha M, Victor VM. Mitochondria, the NLRP3 Inflammasome, and Sirtuins in Type 2 Diabetes: New Therapeutic Targets. Antioxid Redox Signal 2018; 29:749-791. [PMID: 29256638 DOI: 10.1089/ars.2017.7313] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Type 2 diabetes mellitus and hyperglycemia can lead to the development of comorbidities such as atherosclerosis and microvascular/macrovascular complications. Both type 2 diabetes and its complications are related to mitochondrial dysfunction and oxidative stress. Type 2 diabetes is also a chronic inflammatory condition that leads to inflammasome activation and the release of proinflammatory mediators, including interleukins (ILs) IL-1β and IL-18. Moreover, sirtuins are energetic sensors that respond to metabolic load, which highlights their relevance in metabolic diseases, such as type 2 diabetes. Recent Advances: Over the past decade, great progress has been made in clarifying the signaling events regulated by mitochondria, inflammasomes, and sirtuins. Nod-like receptor family pyrin domain containing 3 (NLRP3) is the best characterized inflammasome, and the generation of oxidant species seems to be critical for its activation. NLRP3 inflammasome activation and altered sirtuin levels have been observed in type 2 diabetes. Critical Issue: Despite increasing evidence of the relationship between the NLRP3 inflammasome, mitochondrial dysfunction, and oxidative stress and of their participation in type 2 diabetes physiopathology, therapeutic strategies to combat type 2 diabetes that target NLRP3 inflammasome and sirtuins are yet to be consolidated. FUTURE DIRECTIONS In this review article, we attempt to provide an overview of the existing literature concerning the crosstalk between mitochondrial impairment and the inflammasome, with particular attention to cellular and mitochondrial redox metabolism and the potential role of the NLRP3 inflammasome and sirtuins in the pathogenesis of type 2 diabetes. In addition, we discuss potential targets for therapeutic intervention based on these molecular interactions. Antioxid. Redox Signal. 29, 749-791.
Collapse
Affiliation(s)
- Susana Rovira-Llopis
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Nadezda Apostolova
- 2 Department of Pharmacology, University of Valencia , Valencia, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Celia Bañuls
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Jordi Muntané
- 3 Department of General Surgery, Hospital University "Virgen del Rocío"/IBiS/CSIC/University of Seville , Seville, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Milagros Rocha
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| | - Victor M Victor
- 1 Service of Endocrinology and Nutrition, University Hospital Doctor Peset , Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- 4 CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
- 5 Department of Physiology, University of Valencia , Valencia, Spain
| |
Collapse
|
33
|
Zepeda-Cervantes J, Vaca L. Induction of adaptive immune response by self-aggregating peptides. Expert Rev Vaccines 2018; 17:723-738. [PMID: 30074424 DOI: 10.1080/14760584.2018.1507742] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Recently, subunit vaccines are replacing some of the traditional vaccines because they offer a higher margin of safety. However, generally subunit vaccines have low antigenicity. Adjuvants are used in vaccine formulations to increase their immunogenicity, but current research suggests that adjuvants could induce serious side effects in susceptible individuals; therefore, the improvement of antigens and adjuvants is important. AREAS COVERED Here we reviewed some self-aggregating peptides (SAPs) used as antigen delivery systems. SAPs are based on a short sequence of amino acids, which have self-aggregating properties, inducing self-interaction among peptide molecules by means of non-covalent interactions to generate nanoparticles (NPs). EXPERT COMMENTARY SAPs increase the immunogenicity of fused/conjugated antigens because they can interact with antigen-presenting cells and induce adaptive immunity based on both humoral and cellular responses. As an example, we report an antigen delivery system based on SAPs forming NPs. These NPs are synthesized using a recombinant baculovirus. We fused the green fluorescent protein to the first 110 amino acids of polyhedrin protein from Autographa californica nucleopolyhedrovirus, which has self-aggregating properties. We showed that these NPs prompt high antibody levels without inducing inflammation, similarly to some SAPs reported here.
Collapse
Affiliation(s)
- Jesus Zepeda-Cervantes
- a Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad Universitaria, CDMX , Coyoacán , Mexico
| | - Luis Vaca
- a Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad Universitaria, CDMX , Coyoacán , Mexico
| |
Collapse
|
34
|
Saeed M. Locus and gene-based GWAS meta-analysis identifies new diabetic nephropathy genes. Immunogenetics 2018; 70:347-353. [PMID: 29147756 DOI: 10.1007/s00251-017-1044-0/tables/2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 11/02/2017] [Indexed: 05/22/2023]
Abstract
Objective Assimilation of SNPs Interacting in Synchrony (OASIS) is a locus-based clustering algorithm recently described that can potentially address false positives and negatives in genome-wide association studies (GWAS) of complex disorders. Diabetic nephropathy (DN) is incompletely understood due to a paucity of genes identified despite several GWAS. OASIS was applied to three DN dbGAP GWAS datasets (4725 subjects; 1.06 million SNPs). OASIS identified 19 DN genes which were verified using single variant replication in a standard association study and gene-based analysis using GATES. CARS and FRMD3 were confirmed as DN genes, and five known diabetes-associated genes, viz. NLRP3, INPPL1, PIK3C2G, NRXN3, and TBC1D4, not previously identified using these datasets were discovered. Furthermore, three additional novel DN genes were found which replicated in two sets of analysis, viz. NTN1, EBF2, and DNAH11. Hence, composite analysis with OASIS, gene-based, and single variant association testing can be universally applied to existing GWAS datasets for the identification of new genes.
Collapse
Affiliation(s)
- Mohammad Saeed
- Department of Genomics, Arkana Laboratories, 10810 Executive Center Drive, Suite 100, Little Rock, AR, 72211, USA.
| |
Collapse
|
35
|
Roncero-Ramos I, Rangel-Zuñiga OA, Lopez-Moreno J, Alcala-Diaz JF, Perez-Martinez P, Jimenez-Lucena R, Castaño JP, Roche HM, Delgado-Lista J, Ordovas JM, Camargo A, Lopez-Miranda J. Mediterranean Diet, Glucose Homeostasis, and Inflammasome Genetic Variants: The CORDIOPREV Study. Mol Nutr Food Res 2018; 62:e1700960. [PMID: 29573224 DOI: 10.1002/mnfr.201700960] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/17/2018] [Indexed: 12/16/2022]
Abstract
SCOPE Insulin resistance (IR) and chronic low-grade inflammation are hallmarks of type 2 diabetes mellitus (T2DM). The "NOD-like receptor pyrin domain containing-3" (NLRP3) inflammasome component of innate immunity is a metabolic stress sensor modulated by dietary and genetics factors. The aim of this study was to evaluate the effects of the consumption of two diets for 3 years, Mediterranean (Med) and low fat, on glucose homeostasis in the 1002 coronary heart disease patients of the CORDIOPREV study, according to a genetic variant of NLRP3 inflammasome. METHODS AND RESULTS The study was conducted in the framework of the CORDIOPREV study, a randomized dietary intervention with Med and low-fat diets. Single nucleotide polymorphisms (SNPs) located at inflammasome NLRP3 gene were genotyped by OpenArray platform. Nondiabetic CT+TT carriers of the rs4612666 SNP and AG+AA carriers of the rs10733113 SNP increased insulin sensitivity index (ISI) after 3 years of dietary intervention, whereas no effect was observed in diabetic patients. Further analysis by diet showed that the improvement of the ISI in nondiabetic rs10733113 AG+AA carriers was specific to the consumption of the Med diet. CONCLUSION Our results show that the benefits associated with a Med diet regarding glucose homeostasis in non-T2DM patients depend on genetic variation in the inflammasome.
Collapse
Affiliation(s)
- Irene Roncero-Ramos
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Oriol A Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Javier Lopez-Moreno
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Rosa Jimenez-Lucena
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Justo P Castaño
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain.,Department of Cell Biology, Physiology, and Immunology, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain
| | - Helen M Roche
- Nutrigenomics Research Group, UCD Conway Institute of Biomolecular Research, and School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin 4, Ireland
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA.,Madrid Institute of Advanced Studies (IMDEA) Alimentacion, Madrid, 28049, Spain.,Spanish National Centre for Cardiovascular Research (CNIC), Madrid, 28029, Spain
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, GC9 Nutrigenomics, Institute Maimonides for Biomedical Research of Cordoba, Reina Sofia University Hospital, University of Cordoba, Cordoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, 14004, Spain
| |
Collapse
|
36
|
Sharma A, Tate M, Mathew G, Vince JE, Ritchie RH, de Haan JB. Oxidative Stress and NLRP3-Inflammasome Activity as Significant Drivers of Diabetic Cardiovascular Complications: Therapeutic Implications. Front Physiol 2018. [PMID: 29515457 PMCID: PMC5826188 DOI: 10.3389/fphys.2018.00114] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
It is now increasingly appreciated that inflammation is not limited to the control of pathogens by the host, but rather that sterile inflammation which occurs in the absence of viral or bacterial pathogens, accompanies numerous disease states, none more so than the complications that arise as a result of hyperglycaemia. Individuals with type 1 or type 2 diabetes mellitus (T1D, T2D) are at increased risk of developing cardiac and vascular complications. Glucose and blood pressure lowering therapies have not stopped the advance of these morbidities that often lead to fatal heart attacks and/or stroke. A unifying mechanism of hyperglycemia-induced cellular damage was initially proposed to link elevated blood glucose levels with oxidative stress and the dysregulation of metabolic pathways. Pre-clinical evidence has, in most cases, supported this notion. However, therapeutic strategies to lessen oxidative stress in clinical trials has not proved efficacious, most likely due to indiscriminate targeting by antioxidants such as vitamins. Recent evidence now suggests that oxidative stress is a major driver of inflammation and vice versa, with the latest findings suggesting not only a key role for inflammatory pathways underpinning metabolic and haemodynamic dysfunction in diabetes, but furthermore that these perturbations are driven by activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. This review will address these latest findings with an aim of highlighting the interconnectivity between oxidative stress, NLRP3 activation and inflammation as it pertains to cardiac and vascular injury sustained by diabetes. Current therapeutic strategies to lessen both oxidative stress and inflammation will be emphasized. This will be placed in the context of improving the burden of these diabetic complications.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Oxidative Stress Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mitchel Tate
- Heart Failure Pharmacology Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geetha Mathew
- Cellular Therapies Laboratory, Westmead Hospital, Sydney, NSW, Australia
| | - James E Vince
- Inflammation Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Judy B de Haan
- Oxidative Stress Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Boff D, Fagundes CT, Russo RC, Amaral FA. Innate Immunity and Inflammation: The Molecular Mechanisms Governing the Cross-Talk Between Innate Immune and Endothelial Cells. IMMUNOPHARMACOLOGY AND INFLAMMATION 2018:33-56. [DOI: 10.1007/978-3-319-77658-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
38
|
Saeed M. Locus and gene-based GWAS meta-analysis identifies new diabetic nephropathy genes. Immunogenetics 2017; 70:347-353. [PMID: 29147756 DOI: 10.1007/s00251-017-1044-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022]
Abstract
Objective Assimilation of SNPs Interacting in Synchrony (OASIS) is a locus-based clustering algorithm recently described that can potentially address false positives and negatives in genome-wide association studies (GWAS) of complex disorders. Diabetic nephropathy (DN) is incompletely understood due to a paucity of genes identified despite several GWAS. OASIS was applied to three DN dbGAP GWAS datasets (4725 subjects; 1.06 million SNPs). OASIS identified 19 DN genes which were verified using single variant replication in a standard association study and gene-based analysis using GATES. CARS and FRMD3 were confirmed as DN genes, and five known diabetes-associated genes, viz. NLRP3, INPPL1, PIK3C2G, NRXN3, and TBC1D4, not previously identified using these datasets were discovered. Furthermore, three additional novel DN genes were found which replicated in two sets of analysis, viz. NTN1, EBF2, and DNAH11. Hence, composite analysis with OASIS, gene-based, and single variant association testing can be universally applied to existing GWAS datasets for the identification of new genes.
Collapse
Affiliation(s)
- Mohammad Saeed
- Department of Genomics, Arkana Laboratories, 10810 Executive Center Drive, Suite 100, Little Rock, AR, 72211, USA.
| |
Collapse
|
39
|
Zhang X, Dai J, Li L, Chen H, Chai Y. NLRP3 Inflammasome Expression and Signaling in Human Diabetic Wounds and in High Glucose Induced Macrophages. J Diabetes Res 2017; 2017:5281358. [PMID: 28164132 PMCID: PMC5259616 DOI: 10.1155/2017/5281358] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/27/2016] [Accepted: 12/06/2016] [Indexed: 11/18/2022] Open
Abstract
Introduction. To investigate the contribution and mechanism of NLRP3 inflammasome expression in human wounds in diabetes mellitus and in high glucose induced macrophages. Methods. In the present study, we compared the expression of NLRP3 inflammasome in debridement wound tissue from diabetic and nondiabetic patients. We also examined whether high glucose induces NLRP3 inflammasome expression in cultures THP-1-derived macrophages and the influence on IL-1β expression. Results. The expressions of NLRP3, caspase1, and IL-1β, at both the mRNA and protein level, were significantly higher in wounds of diabetic patients compared with nondiabetic wounds (P < 0.05). High glucose induced a significant increase in NLRP3 inflammasome and IL-1β expression in THP-1-derived macrophages. M1 macrophage surface marker with CCR7 was significantly upregulated after high glucose stimulation. SiRNA-mediated silencing of NLRP3 expression downregulates the expression of IL-1β. Conclusion. The higher expression of NLRP3, caspase1, and secretion of IL-1β, signaling, and activation might contribute to the hyperinflammation in the human diabetic wound and in high glucose induced macrophages. It may be a novel target to treat the DM patients with chronic wound.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Jiao Tong University, Shanghai, China
| | - Jiezhi Dai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Jiao Tong University, Shanghai, China
| | - Li Li
- Department of Orthopedic Surgery, Shanghai First People's Hospital, Jiao Tong University, Shanghai, China
| | - Hua Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Jiao Tong University, Shanghai, China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Jiao Tong University, Shanghai, China
| |
Collapse
|
40
|
Hardigan T, Ward R, Ergul A. Cerebrovascular complications of diabetes: focus on cognitive dysfunction. Clin Sci (Lond) 2016; 130:1807-22. [PMID: 27634842 PMCID: PMC5599301 DOI: 10.1042/cs20160397] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/11/2015] [Indexed: 01/01/2023]
Abstract
The incidence of diabetes has more than doubled in the United States in the last 30 years and the global disease rate is projected to double by 2030. Cognitive impairment has been associated with diabetes, worsening quality of life in patients. The structural and functional interaction of neurons with the surrounding vasculature is critical for proper function of the central nervous system including domains involved in learning and memory. Thus, in this review we explore cognitive impairment in patients and experimental models, focusing on links to vascular dysfunction and structural changes. Lastly, we propose a role for the innate immunity-mediated inflammation in neurovascular changes in diabetes.
Collapse
Affiliation(s)
- Trevor Hardigan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, U.S.A
| | - Rebecca Ward
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, U.S.A
| | - Adviye Ergul
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, U.S.A. Charlie Norwood Veterans Administration Medical Center, Augusta, GA 30912, U.S.A.
| |
Collapse
|
41
|
Lénárt N, Brough D, Dénes Á. Inflammasomes link vascular disease with neuroinflammation and brain disorders. J Cereb Blood Flow Metab 2016; 36:1668-1685. [PMID: 27486046 PMCID: PMC5076791 DOI: 10.1177/0271678x16662043] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022]
Abstract
The role of inflammation in neurological disorders is increasingly recognised. Inflammatory processes are associated with the aetiology and clinical progression of migraine, psychiatric conditions, epilepsy, cerebrovascular diseases, dementia and neurodegeneration, such as seen in Alzheimer's or Parkinson's disease. Both central and systemic inflammatory actions have been linked with the development of brain diseases, suggesting that complex neuro-immune interactions could contribute to pathological changes in the brain across multiple temporal and spatial scales. However, the mechanisms through which inflammation impacts on neurological disease are improperly defined. To develop effective therapeutic approaches, it is imperative to understand how detrimental inflammatory processes could be blocked selectively, or controlled for prolonged periods, without compromising essential immune defence mechanisms. Increasing evidence indicates that common risk factors for brain disorders, such as atherosclerosis, diabetes, hypertension, obesity or infection involve the activation of NLRP3, NLRP1, NLRC4 or AIM2 inflammasomes, which are also associated with various neurological diseases. This review focuses on the mechanisms whereby inflammasomes, which integrate diverse inflammatory signals in response to pathogen-driven stimuli, tissue injury or metabolic alterations in multiple cell types and different organs of the body, could functionally link vascular- and neurological diseases and hence represent a promising therapeutic target.
Collapse
Affiliation(s)
- Nikolett Lénárt
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - David Brough
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK
| | - Ádám Dénes
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|