1
|
Zhao X, Wu J, Lai J, Pan B, Ji M, Li X, He Y, Han J. CITMIC: Comprehensive Estimation of Cell Infiltration in Tumor Microenvironment based on Individualized Intercellular Crosstalk. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408007. [PMID: 39498855 DOI: 10.1002/advs.202408007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/27/2024] [Indexed: 11/07/2024]
Abstract
The tumor microenvironment (TME) cells interact with each other and play a pivotal role in tumor progression and treatment response. A comprehensive characterization of cell and intercellular crosstalk in the TME is essential for understanding tumor biology and developing effective therapies. However, current cell infiltration analysis methods only partially describe the TME's cellular landscape and overlook cell-cell crosstalk. Here, this approach, CITMIC, can infer the cell infiltration of TME by simultaneously measuring 86 different cell types, constructing an individualized cell-cell crosstalk network based on functional similarities between cells, and using only gene transcription data. This is a novel approach to estimating the relative cell infiltration levels, which are shown to be superior to the current methods. The TME cell-based features generated by analyzing melanoma data are effective in predicting prognosis and treatment response. Interestingly, these features are found to be particularly effective in assessing the prognosis of high-stage patients, and this method is applied to multiple high-stage adenocarcinomas, where more significant prognostic performance is also observed. In conclusion, CITMIC offers a more comprehensive description of TME cell composition by considering cell-cell crosstalk, providing an important reference for the discovery of predictive biomarkers and the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Xilong Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jiashuo Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jiyin Lai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Bingyue Pan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Miao Ji
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xiangmei Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yalan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
2
|
Li X, Zhu Y, Yan T, Fang J, Xu X, Xu X. Association between C-reactive protein, Life's Essential 8, and mortality in American adults: Insights from NHANES 2005-2010 data analysis. Exp Gerontol 2024; 196:112590. [PMID: 39307250 DOI: 10.1016/j.exger.2024.112590] [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: 07/16/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE To evaluate the independent, mediating, interactive, and associated effects of C-reactive protein (CRP) and Life's Essential 8 (LE8) on all-cause and cardiovascular mortality. METHODS Utilizing data from 10,043 participants in the NHANES 2005-2010, we employed Cox proportional hazards regression models and causal mediation analysis to investigate the joint and interactive effects of Life's Essential 8 (LE8) and C-reactive protein (CRP) on mortality risk. RESULTS During an average follow-up of 137.10 months, there were 1591 all-cause deaths and 485 cardiovascular deaths. Weighted linear regression showed that for patients with low cardiovascular health (CVH), the adjusted β was -0.22 (95 % CI: -0.27 to -0.17) for moderate cardiovascular health (CVH) and -0.36 (95 % CI: -0.43 to -0.30) for high cardiovascular health (CVH). Mediation analysis revealed that C-reactive protein (CRP) mediated 10.43 % of all-cause mortality and 9.20 % of cardiovascular mortality for moderate cardiovascular health (CVH) compared to low cardiovascular health (CVH), and 9.95 % and 8.32 % respectively for high cardiovascular health (CVH) compared to low cardiovascular health (CVH). No significant multiplicative or additive interactions between Life's Essential 8 (LE8) and C-reactive protein (CRP) were found in all-cause mortality or cardiovascular mortality. Individuals with high cardiovascular health (CVH) and the first quartile of C-reactive protein (CRP) had HRs of 0.30 (95 % CI 0.18-0.48) for all-cause mortality and 0.31 (95 % CI 0.13-0.74) for cardiovascular mortality compared to those with low cardiovascular health (CVH) and the fourth quartile of C-reactive protein (CRP). CONCLUSION These findings suggest that CRP could significantly influence the relationship between cardiovascular health (CVH) and mortality. Interventions that target both lifestyle factors and CRP levels may improve cardiovascular health and potentially lower mortality risks.
Collapse
Affiliation(s)
- Xin Li
- Department of Cardiovascular Diseases, The People's Hospital of Chizhou, Chizhou 247100, China
| | - Yongxin Zhu
- Department of Cardiovascular Diseases, The People's Hospital of Chizhou, Chizhou 247100, China
| | - Tingting Yan
- Department of Cardiovascular Diseases, The People's Hospital of Chizhou, Chizhou 247100, China
| | - Jie Fang
- Department of Cardiovascular Diseases, The People's Hospital of Chizhou, Chizhou 247100, China
| | - Xin Xu
- Department of Cardiovascular Diseases, The People's Hospital of Chizhou, Chizhou 247100, China
| | - Xiaodong Xu
- Department of Cardiovascular Diseases, The People's Hospital of Chizhou, Chizhou 247100, China.
| |
Collapse
|
3
|
Wei B, Cheng G, Bi Q, Lu C, Sun Q, Li L, Chen N, Hu M, Lu H, Xu X, Mao G, Wan S, Hu Z, Gu Y, Zheng J, Zhao L, Shen XZ, Liu X, Shi P. Microglia in the hypothalamic paraventricular nucleus sense hemodynamic disturbance and promote sympathetic excitation in hypertension. Immunity 2024; 57:2030-2042.e8. [PMID: 39116878 DOI: 10.1016/j.immuni.2024.07.011] [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: 09/06/2023] [Revised: 04/22/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Hypertension is usually accompanied by elevated sympathetic tonicity, but how sympathetic hyperactivity is triggered is not clear. Recent advances revealed that microglia-centered neuroinflammation contributes to sympathetic excitation in hypertension. In this study, we performed a temporospatial analysis of microglia at both morphological and transcriptomic levels and found that microglia in the hypothalamic paraventricular nucleus (PVN), a sympathetic center, were early responders to hypertensive challenges. Vasculature analyses revealed that the PVN was characterized by high capillary density, thin vessel diameter, and complex vascular topology relative to other brain regions. As such, the PVN was susceptible to the penetration of ATP released from the vasculature in response to hemodynamic disturbance after blood pressure increase. Mechanistically, ATP ligation to microglial P2Y12 receptor was responsible for microglial inflammatory activation and the eventual sympathetic overflow. Together, these findings identified a distinct vasculature pattern rendering vulnerability of PVN pre-sympathetic neurons to hypertension-associated microglia-mediated inflammatory insults.
Collapse
Affiliation(s)
- Bo Wei
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Guo Cheng
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Qianqian Bi
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Cheng Lu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Qihang Sun
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Li Li
- Department of Pharmacy, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Ningting Chen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Miner Hu
- Department of Cardiology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Haoran Lu
- Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China
| | - Xuancheng Xu
- Zhejiang Chinese Medical University, Hangzhou 310013, China; Department of Neurology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shu Wan
- Brain Center, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China
| | - Zhechun Hu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China; Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Gu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China; Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxin Zheng
- Key Laboratory for Biomedical Engineering of Ministrey of Education, Collage of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310013, China
| | - Li Zhao
- Key Laboratory for Biomedical Engineering of Ministrey of Education, Collage of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310013, China
| | - Xiao Z Shen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Brain Center, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China; Department of Physiology, Zhejiang University School of Medicine, Hangzhou 310058, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310013, China
| | - Xiaoli Liu
- Department of Neurology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310013, China.
| | - Peng Shi
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310013, China.
| |
Collapse
|
4
|
Woodfin S, Hall S, Ramerth A, Chapple B, Fausnacht D, Moore W, Alkhalidy H, Liu D. Potential Application of Plant-Derived Compounds in Multiple Sclerosis Management. Nutrients 2024; 16:2996. [PMID: 39275311 PMCID: PMC11397714 DOI: 10.3390/nu16172996] [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: 07/30/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by inflammation, demyelination, and neurodegeneration, resulting in significant disability and reduced quality of life. Current therapeutic strategies primarily target immune dysregulation, but limitations in efficacy and tolerability highlight the need for alternative treatments. Plant-derived compounds, including alkaloids, phenylpropanoids, and terpenoids, have demonstrated anti-inflammatory effects in both preclinical and clinical studies. By modulating immune responses and promoting neuroregeneration, these compounds offer potential as novel adjunctive therapies for MS. This review provides insights into the molecular and cellular basis of MS pathogenesis, emphasizing the role of inflammation in disease progression. It critically evaluates emerging evidence supporting the use of plant-derived compounds to attenuate inflammation and MS symptomology. In addition, we provide a comprehensive source of information detailing the known mechanisms of action and assessing the clinical potential of plant-derived compounds in the context of MS pathogenesis, with a focus on their anti-inflammatory and neuroprotective properties.
Collapse
Affiliation(s)
- Seth Woodfin
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Sierra Hall
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Alexis Ramerth
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Brooke Chapple
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Dane Fausnacht
- Department of Biology, School of Sciences and Agriculture, Ferrum College, Ferrum, VA 24088, USA
| | - William Moore
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Hana Alkhalidy
- Department of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
5
|
Sahu Y, Jamadade P, Ch Maharana K, Singh S. Role of mitochondrial homeostasis in D-galactose-induced cardiovascular ageing from bench to bedside. Mitochondrion 2024; 78:101923. [PMID: 38925493 DOI: 10.1016/j.mito.2024.101923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Ageing is an inevitable phenomenon which affects the cellular to the organism level in the progression of the time. Oxidative stress and inflammation are now widely regarded as the key processes involved in the aging process, which may then cause significant harm to mitochondrial DNA, leading to apoptosis. Normal circulatory function is a significant predictor of disease-free life expectancy. Indeed, disorders affecting the cardiovascular system, which are becoming more common, are the primary cause of worldwide morbidity, disability, and mortality. Cardiovascular aging may precede or possibly underpin overall, age-related health decline. Numerous studies have foundmitochondrial mechanistc approachplays a vital role in the in the onset and development of aging. The D-galactose (D-gal)-induced aging model is well recognized and commonly used in the aging study. In this review we redeposit the association of the previous and current studies on mitochondrial homeostasis and its underlying mechanisms in D-galactose cardiovascular ageing. Further we focus the novel and the treatment strategies to combat the major complication leading to the cardiovascular ageing.
Collapse
Affiliation(s)
- Yogita Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Pratiksha Jamadade
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Krushna Ch Maharana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hajipur, Vaishali, Bihar, India.
| |
Collapse
|
6
|
Noh JY, Han HW, Kim DM, Giles ED, Farnell YZ, Wright GA, Sun Y. Innate immunity in peripheral tissues is differentially impaired under normal and endotoxic conditions in aging. Front Immunol 2024; 15:1357444. [PMID: 39221237 PMCID: PMC11361940 DOI: 10.3389/fimmu.2024.1357444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Chronic low-grade inflammation is a hallmark of aging, aka "inflammaging", which is linked to a wide range of age-associated diseases. Immune dysfunction increases disease susceptibility, and increases morbidity and mortality of aging. Innate immune cells, including monocytes, macrophages and neutrophils, are the first responders of host defense and the key mediators of various metabolic and inflammatory insults. Currently, the understanding of innate immune programming in aging is largely fragmented. Here we investigated the phenotypic and functional properties of innate immune cells in various peripheral tissues of young and aged mice under normal and endotoxic conditions. Under the steady state, aged mice showed elevated pro-inflammatory monocytes/macrophages in peripheral blood, adipose tissue, liver, and colon. Under lipopolysaccharide (LPS)-induced inflammatory state, the innate immune cells of aged mice showed a different response to LPS stimulus than that of young mice. LPS-induced immune responses displayed differential profiles in different tissues and cell types. In the peripheral blood, when responding to LPS, the aged mice showed higher neutrophils, but lower pro-inflammatory monocytes than that in young mice. In the peritoneal fluid, while young mice exhibited significantly elevated pro-inflammatory neutrophils and macrophages in response to LPS, aged mice exhibited decreased pro-inflammatory neutrophils and variable cytokine responses in macrophages. In the adipose tissue, LPS induced less infiltrated neutrophils but more infiltrated macrophages in old mice than young mice. In the liver, aged mice showed a more robust increase of pro-inflammatory macrophages compared to that in young mice under LPS stimulation. In colon, macrophages showed relatively mild response to LPS in both young and old mice. We have further tested bone-marrow derived macrophages (BMDM) from young and aged mice, we found that BMDM from aged mice have impaired polarization, displaying higher expression of pro-inflammatory markers than those from young mice. These data collectively suggest that innate immunity in peripheral tissues is impaired in aging, and the dysregulation of immunity is tissue- and cell-dependent. Our findings in the rodent model underscore the complexity of aging immunity. Further investigation is needed to determine whether the immune profile observed in aged mice is applicable in age-associated diseases in humans.
Collapse
Affiliation(s)
- Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States
| | - Yuhua Z. Farnell
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
| | - Gus A. Wright
- Department of Veterinary Pathobiology, Texas A&M University,
College Station, TX, United States
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
7
|
Liu L, Zhu L, Liang Q, Yu L, Hu L, Yu Y, Cheng X, Bao H. Tissue-resident C1q + macrophages exert anti-aging potential through the Sirt1 pathway. Inflamm Res 2024; 73:1069-1080. [PMID: 38724770 DOI: 10.1007/s00011-024-01883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/03/2024] [Accepted: 04/08/2024] [Indexed: 07/01/2024] Open
Abstract
OBJECTIVE Resident immune cells are at the forefront of sensory organ-specific signals, and changes in these cells are closely related to the aging process. The Sirt pathway can regulate NAD + metabolism during aging, thereby affecting the accumulation of ROS. However, the role of the Sirt pathway in resident immune cells in aged tissues is currently unclear. METHODS We investigated Sirt1 signalling in resident immune cells during chronic inflammation in an aged mouse model. Integrated single-cell RNA sequencing data from young and aged mice were used to refine the characterization of immune cells in aged tissues RESULTS: We found that C1q + macrophages could affect chronic inflammation during aging. C1q + macrophages acted in an opposing manner to Il1b + macrophages and were responsible for anti-inflammatory effects during aging. Sirt1 agonists inhibited the decrease in C1qb in macrophages during aging, and anti-aging drugs could affect the expression of C1qb in macrophages via the Sirt1 pathway. CONCLUSIONS In this study, we first identified the relevance of C1q + macrophages in chronic inflammation during aging. The potential anti-aging effect of C1q + macrophages was mediated by the Sirt1 pathway, suggesting new strategies for aging immunotherapy.
Collapse
Affiliation(s)
- Liang Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, 330000, China
- Jiangxi Sub-Center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, 330000, China
| | - Lingjuan Zhu
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, 330000, China
- Jiangxi Sub-Center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, 330000, China
| | - Qian Liang
- Department of Science and Technology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Lingling Yu
- Department of Rehabilitation, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Longlong Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Yun Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Xiaoshu Cheng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, 330000, China.
- Jiangxi Sub-Center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, 330000, China.
| | - Huihui Bao
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, 330000, China.
- Jiangxi Sub-Center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, 330000, China.
| |
Collapse
|
8
|
Thangaraj SS, Oxlund CS, Andersen H, Svenningsen P, Stubbe J, Palarasah Y, Fonseca MPD, Ketelhuth DFJ, Enggaard C, Hansen MH, Henriksen JE, Jacobsen IA, Jensen BL. Amiloride lowers plasma TNF and interleukin-6 but not interleukin-17A in patients with hypertension and type 2 diabetes. Am J Physiol Renal Physiol 2024; 327:F37-F48. [PMID: 38779752 DOI: 10.1152/ajprenal.00268.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 04/12/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Interleukin (IL)-17A contributes to hypertension in preclinical models. T helper 17 and dendritic cells are activated by NaCl, which could involve the epithelial Na+ channel (ENaC). We hypothesized that the ENaC blocker amiloride reduces plasma IL-17A and related cytokines in patients with hypertension. Concentrations of IL-17A, IFN-γ, TNF, IL-6, IL-1β, and IL-10 were determined by immunoassays in plasma from two patient cohorts before and after amiloride treatment: 1) patients with type 2 diabetes mellitus (T2DM) and treatment-resistant hypertension (n = 69, amiloride 5-10 mg/day for 8 wk) and 2) patients with hypertension and type 1 diabetes mellitus (T1DM) (n = 29) on standardized salt intake (amiloride 20-40 mg/day, 2 days). Plasma and tissue from ANG II-hypertensive mice with T1DM treated with amiloride (2 mg/kg/day, 4 days) were analyzed. The effect of amiloride and benzamil on macrophage cytokines was determined in vitro. Plasma cytokines showed higher concentrations (IL-17A ∼40-fold) in patients with T2DM compared with T1DM. In patients with T2DM, amiloride had no effect on IL-17A but lowered TNF and IL-6. In patients with T1DM, amiloride had no effect on IL-17A but increased TNF. In both cohorts, blood pressure decline and plasma K+ increase did not relate to plasma cytokine changes. In mice, amiloride exerted no effect on IL-17A in the plasma, kidney, aorta, or left cardiac ventricle but increased TNF in cardiac and kidney tissues. In lipopolysaccharide-stimulated human THP-1 macrophages, amiloride and benzamil (from 1 nmol/L) decreased TNF, IL-6, IL-10, and IL-1β. In conclusion, inhibition of ENaC by amiloride reduces proinflammatory cytokines TNF and IL-6 but not IL-17A in patients with T2DM, potentially by a direct action on macrophages.NEW & NOTEWORTHY ENaC activity may contribute to macrophage-derived cytokine release, since amiloride exerts anti-inflammatory effects by suppression of TNF and IL-6 cytokines in patients with resistant hypertension and type 2 diabetes and in THP-1-derived macrophages in vitro.
Collapse
Affiliation(s)
- Sai Sindhu Thangaraj
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Christina S Oxlund
- Department of Cardiology, Hospital of Southwest Jutland, Esbjerg, Denmark
| | - Henrik Andersen
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Per Svenningsen
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jane Stubbe
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Yaseelan Palarasah
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Micaella Pereira Da Fonseca
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Daniel F J Ketelhuth
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Camilla Enggaard
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Maria Høj Hansen
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Ib Abildgaard Jacobsen
- Research Unit for Cardiovascular and Metabolic Prevention, Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
9
|
Al-Turki TM, Maranon DG, Nelson CB, Lewis AM, Luxton JJ, Taylor LE, Altina N, Wu F, Du H, Kim J, Damle N, Overbey E, Meydan C, Grigorev K, Winer DA, Furman D, Mason CE, Bailey SM. Telomeric RNA (TERRA) increases in response to spaceflight and high-altitude climbing. Commun Biol 2024; 7:698. [PMID: 38862827 PMCID: PMC11167063 DOI: 10.1038/s42003-024-06014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/06/2024] [Indexed: 06/13/2024] Open
Abstract
Telomeres are repetitive nucleoprotein complexes at chromosomal termini essential for maintaining genome stability. Telomeric RNA, or TERRA, is a previously presumed long noncoding RNA of heterogeneous lengths that contributes to end-capping structure and function, and facilitates telomeric recombination in tumors that maintain telomere length via the telomerase-independent Alternative Lengthening of Telomeres (ALT) pathway. Here, we investigated TERRA in the radiation-induced DNA damage response (DDR) across astronauts, high-altitude climbers, healthy donors, and cellular models. Similar to astronauts in the space radiation environment and climbers of Mt. Everest, in vitro radiation exposure prompted increased transcription of TERRA, while simulated microgravity did not. Data suggest a specific TERRA DDR to telomeric double-strand breaks (DSBs), and provide direct demonstration of hybridized TERRA at telomere-specific DSB sites, indicative of protective TERRA:telomeric DNA hybrid formation. Targeted telomeric DSBs also resulted in accumulation of TERRA foci in G2-phase, supportive of TERRA's role in facilitating recombination-mediated telomere elongation. Results have important implications for scenarios involving persistent telomeric DNA damage, such as those associated with chronic oxidative stress (e.g., aging, systemic inflammation, environmental and occupational radiation exposures), which can trigger transient ALT in normal human cells, as well as for targeting TERRA as a therapeutic strategy against ALT-positive tumors.
Collapse
Affiliation(s)
- Taghreed M Al-Turki
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
- Lineberger Comprehensive Cancer Center and Departments of Microbiology and Immunology, and Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - David G Maranon
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Christopher B Nelson
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
- Children's Medical Research Institute, 214 Hawkesbury Road, Westmead, Sydney, NSW, 2145, Australia
| | - Aidan M Lewis
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Jared J Luxton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Lynn E Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Noelia Altina
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Fei Wu
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, USA
| | - Huixun Du
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Eliah Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Kirill Grigorev
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Daniel A Winer
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, USA
| | - David Furman
- Buck AI Platform, Buck Institute for Research on Aging, Novato, CA, USA
- Stanford 1000 Immunomes Project, Stanford School of Medicine, Stanford, CA, USA
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, CONICET, Pilar, Argentina
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA.
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
10
|
Singh A, Schurman SH, Bektas A, Kaileh M, Roy R, Wilson DM, Sen R, Ferrucci L. Aging and Inflammation. Cold Spring Harb Perspect Med 2024; 14:a041197. [PMID: 38052484 PMCID: PMC11146314 DOI: 10.1101/cshperspect.a041197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Aging can be conceptualized as the progressive disequilibrium between stochastic damage accumulation and resilience mechanisms that continuously repair that damage, which eventually cause the development of chronic disease, frailty, and death. The immune system is at the forefront of these resilience mechanisms. Indeed, aging is associated with persistent activation of the immune system, witnessed by a high circulating level of inflammatory markers and activation of immune cells in the circulation and in tissue, a condition called "inflammaging." Like aging, inflammaging is associated with increased risk of many age-related pathologies and disabilities, as well as frailty and death. Herein we discuss recent advances in the understanding of the mechanisms leading to inflammaging and the intrinsic dysregulation of the immune function that occurs with aging. We focus on the underlying mechanisms of chronic inflammation, in particular the role of NF-κB and recent studies targeting proinflammatory mediators. We further explore the dysregulation of the immune response with age and immunosenescence as an important mechanistic immune response to acute stressors. We examine the role of the gastrointestinal microbiome, age-related dysbiosis, and the integrated stress response in modulating the inflammatory "response" to damage accumulation and stress. We conclude by focusing on the seminal question of whether reducing inflammation is useful and the results of related clinical trials. In summary, we propose that inflammation may be viewed both as a clinical biomarker of the failure of resilience mechanisms and as a causal factor in the rising burden of disease and disabilities with aging. The fact that inflammation can be reduced through nonpharmacological interventions such as diet and exercise suggests that a life course approach based on education may be a successful strategy to increase the health span with few adverse consequences.
Collapse
Affiliation(s)
- Amit Singh
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Shepherd H Schurman
- Clinical Research Unit, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Arsun Bektas
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Mary Kaileh
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Roshni Roy
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - David M Wilson
- Biomedical Research Institute, Hasselt University, Diepenbeek 3500, Belgium
| | - Ranjan Sen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland 21224, USA
| |
Collapse
|
11
|
Rios FJ, de Ciuceis C, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Lopreiato M, Mavraganis G, Mengozzi A, Montezano AC, Stavropoulos K, Winklewski PJ, Wolf J, Costantino S, Doumas M, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Virdis A. Mechanisms of Vascular Inflammation and Potential Therapeutic Targets: A Position Paper From the ESH Working Group on Small Arteries. Hypertension 2024; 81:1218-1232. [PMID: 38511317 DOI: 10.1161/hypertensionaha.123.22483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Inflammatory responses in small vessels play an important role in the development of cardiovascular diseases, including hypertension, stroke, and small vessel disease. This involves various complex molecular processes including oxidative stress, inflammasome activation, immune-mediated responses, and protein misfolding, which together contribute to microvascular damage. In addition, epigenetic factors, including DNA methylation, histone modifications, and microRNAs influence vascular inflammation and injury. These phenomena may be acquired during the aging process or due to environmental factors. Activation of proinflammatory signaling pathways and molecular events induce low-grade and chronic inflammation with consequent cardiovascular damage. Identifying mechanism-specific targets might provide opportunities in the development of novel therapeutic approaches. Monoclonal antibodies targeting inflammatory cytokines and epigenetic drugs, show promise in reducing microvascular inflammation and associated cardiovascular diseases. In this article, we provide a comprehensive discussion of the complex mechanisms underlying microvascular inflammation and offer insights into innovative therapeutic strategies that may ameliorate vascular injury in cardiovascular disease.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - George Pavlidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
| | - Claudia Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Marcin Hellmann
- Cardiac Diagnostics (M.H.), Medical University of Gdansk, Poland
| | - Stefano Masi
- Institute of Cardiovascular Science, University College London, United Kingdom (S.M.)
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Mariarosaria Lopreiato
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa (A.M.)
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Konstantinos Stavropoulos
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Greece (K. Stavropoulos)
| | - Pawel J Winklewski
- Departments of Human Physiology (P.J.W.), Medical University of Gdansk, Poland
| | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Sarah Costantino
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
| | - Michael Doumas
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - Ignatios Ikonomidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
- Department of Research and Education (F.P.), University Hospital Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
- Division of Medicine, Spedali Civili di Brescia, Italy (D.R.)
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim (K. Stellos), Heidelberg University, Germany
- Department of Cardiology, University Hospital Mannheim (K. Stellos), Heidelberg University, Germany
- German Centre for Cardiovascular Research, Heidelberg/Mannheim Partner Site (K. Stellos)
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| |
Collapse
|
12
|
Guzik TJ, Nosalski R, Maffia P, Drummond GR. Immune and inflammatory mechanisms in hypertension. Nat Rev Cardiol 2024; 21:396-416. [PMID: 38172242 DOI: 10.1038/s41569-023-00964-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Hypertension is a global health problem, with >1.3 billion individuals with high blood pressure worldwide. In this Review, we present an inflammatory paradigm for hypertension, emphasizing the crucial roles of immune cells, cytokines and chemokines in disease initiation and progression. T cells, monocytes, macrophages, dendritic cells, B cells and natural killer cells are all implicated in hypertension. Neoantigens, the NLRP3 inflammasome and increased sympathetic outflow, as well as cytokines (including IL-6, IL-7, IL-15, IL-18 and IL-21) and a high-salt environment, can contribute to immune activation in hypertension. The activated immune cells migrate to target organs such as arteries (especially the perivascular fat and adventitia), kidneys, the heart and the brain, where they release effector cytokines that elevate blood pressure and cause vascular remodelling, renal damage, cardiac hypertrophy, cognitive impairment and dementia. IL-17 secreted by CD4+ T helper 17 cells and γδ T cells, and interferon-γ and tumour necrosis factor secreted by immunosenescent CD8+ T cells, exert crucial effector roles in hypertension, whereas IL-10 and regulatory T cells are protective. Effector mediators impair nitric oxide bioavailability, leading to endothelial dysfunction and increased vascular contractility. Inflammatory effector mediators also alter renal sodium and water balance and promote renal fibrosis. These mechanisms link hypertension with obesity, autoimmunity, periodontitis and COVID-19. A comprehensive understanding of the immune and inflammatory mechanisms of hypertension is crucial for safely and effectively translating the findings to clinical practice.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK.
- Department of Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University, Collegium Medicum, Kraków, Poland.
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK.
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Pasquale Maffia
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Wang M, Guo W, Chen JF. Caffeine: a potential mechanism for anti-obesity. Purinergic Signal 2024:10.1007/s11302-024-10022-1. [PMID: 38802651 DOI: 10.1007/s11302-024-10022-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Obesity refers to the excessive accumulation of fat caused by a long-term imbalance between energy intake (EI) and energy expenditure (EE). Over recent years, obesity has become a major public health challenge. Caffeine is a natural product that has been demonstrated to exert anti-obesity effects; however, the mechanisms responsible for the effect of caffeine on weight loss have yet to be fully elucidated. Most obesity-related deaths are due to cardiovascular disease. Recent research has demonstrated that caffeine can reduce the risk of death from cardiovascular disease; thus, it can be hypothesized that caffeine may represent a new therapeutic agent for weight loss. In this review, we synthesize data arising from clinical and animal studies over the last decade and discuss the potential mechanisms by which caffeine may induce weight loss, focusing particularly on increasing energy consumption, suppressing appetite, altering lipid metabolism, and influencing the gut microbiota. Finally, we summarize the major challenges associated with caffeine and anti-obesity research and highlight possible directions for future research and development.
Collapse
Affiliation(s)
- Meng Wang
- International Joint Research Center on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wei Guo
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
14
|
Vilardi A, Przyborski S, Mobbs C, Rufini A, Tufarelli C. Current understanding of the interplay between extracellular matrix remodelling and gut permeability in health and disease. Cell Death Discov 2024; 10:258. [PMID: 38802341 PMCID: PMC11130177 DOI: 10.1038/s41420-024-02015-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
The intestinal wall represents an interactive network regulated by the intestinal epithelium, extracellular matrix (ECM) and mesenchymal compartment. Under healthy physiological conditions, the epithelium undergoes constant renewal and forms an integral and selective barrier. Following damage, the healthy epithelium is restored via a series of signalling pathways that result in remodelling of the scaffolding tissue through finely-regulated proteolysis of the ECM by proteases such as matrix metalloproteinases (MMPs). However, chronic inflammation of the gastrointestinal tract, as occurs in Inflammatory Bowel Disease (IBD), is associated with prolonged disruption of the epithelial barrier and persistent damage to the intestinal mucosa. Increased barrier permeability exhibits distinctive signatures of inflammatory, immunological and ECM components, accompanied by increased ECM proteolytic activity. This narrative review aims to bring together the current knowledge of the interplay between gut barrier, immune and ECM features in health and disease, discussing the role of barrier permeability as a discriminant between homoeostasis and IBD.
Collapse
Affiliation(s)
- Aurora Vilardi
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Claire Mobbs
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Alessandro Rufini
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
- Department of Biosciences, University of Milan, Milan, 20133, Italy.
| | - Cristina Tufarelli
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
| |
Collapse
|
15
|
Thirugnanam S, Rout N. A Perfect Storm: The Convergence of Aging, Human Immunodeficiency Virus Infection, and Inflammasome Dysregulation. Curr Issues Mol Biol 2024; 46:4768-4786. [PMID: 38785555 PMCID: PMC11119826 DOI: 10.3390/cimb46050287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
The emergence of combination antiretroviral therapy (cART) has greatly transformed the life expectancy of people living with HIV (PWH). Today, over 76% of the individuals with HIV have access to this life-saving therapy. However, this progress has come with a new challenge: an increase in age-related non-AIDS conditions among patients with HIV. These conditions manifest earlier in PWH than in uninfected individuals, accelerating the aging process. Like PWH, the uninfected aging population experiences immunosenescence marked by an increased proinflammatory environment. This phenomenon is linked to chronic inflammation, driven in part by cellular structures called inflammasomes. Inflammatory signaling pathways activated by HIV-1 infection play a key role in inflammasome formation, suggesting a crucial link between HIV and a chronic inflammatory state. This review outlines the inflammatory processes triggered by HIV-1 infection and aging, with a focus on the inflammasomes. This review also explores current research regarding inflammasomes and potential strategies for targeting inflammasomes to mitigate inflammation. Further research on inflammasome signaling presents a unique opportunity to develop targeted interventions and innovative therapeutic modalities for combating HIV and aging-associated inflammatory processes.
Collapse
Affiliation(s)
- Siva Thirugnanam
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Namita Rout
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
16
|
Macech M, Grochowiecki T, Durlik M, Pączek L, Nazarewski S. Impact of Pretransplant C-Reactive Protein, Neutrophiles, Platelets, and Albumin Levels on Recipient Survival After Simultaneous Pancreas and Kidney Transplantation. Transplant Proc 2024; 56:806-812. [PMID: 38729832 DOI: 10.1016/j.transproceed.2024.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/29/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Long-lasting diabetes mellitus type 1 and end-stage renal disease induce severe metabolic and immunologic deterioration. Pretransplant C-reactive protein (CRP) and albumin (ALB) levels impact kidney transplantation. We evaluated the effects of preoperative CRP, ALB, neutrophils (NEU), and platelet (PLT) counts on 1- and 5-year recipient survival after simultaneous pancreas and kidney transplantation (SPK). METHODS Among 103 SPK recipients, the parameters were as follows: CRP (mean: 4.5 ± 4.97 mg/L); NEU (mean: 5.12 ± 2.13 × 103/mm3); PLT (mean: 244 ± 84 × 103/mm3); ALB (mean 4.5 ± 0.75 g/dL) were obtained before transplantation. Cox regression, uni-, multivariate analysis for 1- and 5-year survivals were performed with 95% CIs, and the area under the receiver operating characteristic (ROC) curve (AUC) was assessed. RESULTS In Cox regression, ALB <3.65 g/dL significantly affected 1- and 5-year survivors with hazard ratios of 8 (95% CI, 1.5-38.28; P < .05) and 3.13 (95% CI, 1.45-6.73; P < .05), respectively. In univariate analysis, we found significantly decreased 1-year survival when PLT <180×103/mm3, ALB <3.65 g/dL, NEU >5.8×103/mm3 and CRP >2.25 mg/L with odds ratios (OR) of 6.75 (95% CI, 2.12-21.15); 4.05 (95% CI, 1.3-12.09); 2.97 (95% CI, 1.02-8.64) and 5.51 (95% CI, 1.67-18.19), respectively. Independent factors for 5-year survival were CRP, ALB, and PLT with OR of 4.72 (95% CI, 1.67-13.29), 3.31 (95% CI, 1.18-9.25), and 4.2 (95% CI, 1.39-12.68), respectively. In multivariate analysis, we built 2 models for 1-year survival. Model 1 (ALB+PLT) with ORs of 3.12 (95% CI, 0.97-10.07) and 5.55 (95% CI, 1.67-18.4); and model 2 (CRP+PLT) with ORs of 5.51 (95% CI, 1.5-17.3) and 4.3 (95% CI, 1.2-15.06), respectively. The AUC for models 1 and 2 were 0.74 and 0.759, respectively. CONCLUSIONS NEU, PLT, ALB, and CRP levels assessed before transplantation are independent factors for 1- and 5-year SPK recipient survival.
Collapse
Affiliation(s)
- Michał Macech
- Department of General, Vascular, Endocrine and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland.
| | - Tadeusz Grochowiecki
- Department of General, Vascular, Endocrine and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Durlik
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Sławomir Nazarewski
- Department of General, Vascular, Endocrine and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
17
|
Pang J, Kuang TD, Yu XY, Novák P, Long Y, Liu M, Deng WQ, Zhu X, Yin K. N6-methyladenosine in myeloid cells: a novel regulatory factor for inflammation-related diseases. J Physiol Biochem 2024; 80:249-260. [PMID: 38158555 DOI: 10.1007/s13105-023-01002-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
N6-methyladenosine (m6A) is one of the most abundant epitranscriptomic modifications on eukaryotic mRNA. Evidence has highlighted that m6A is altered in response to inflammation-related factors and it is closely associated with various inflammation-related diseases. Multiple subpopulations of myeloid cells, such as macrophages, dendritic cells, and granulocytes, are crucial for the regulating of immune process in inflammation-related diseases. Recent studies have revealed that m6A plays an important regulatory role in the functional of multiple myeloid cells. In this review, we comprehensively summarize the function of m6A modification in myeloid cells from the perspective of myeloid cell production, activation, polarization, and migration. Furthermore, we discuss how m6A-mediated myeloid cell function affects the progression of inflammation-related diseases, including autoimmune diseases, chronic metabolic diseases, and malignant tumors. Finally, we discuss the challenges encountered in the study of m6A in myeloid cells, intended to provide a new direction for the study of the pathogenesis of inflammation-related diseases.
Collapse
Affiliation(s)
- Jin Pang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Tong-Dong Kuang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Xin-Yuan Yu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Yuan Long
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Min Liu
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Wei-Qian Deng
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China.
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Zhu CS, Chen W, Qiang X, Lou L, Li J, Wang H. Elevated Circulating Procathepsin L as a Potential Biomarker of Inflamm-aging. Med Hypotheses 2024; 186:111322. [PMID: 38617026 PMCID: PMC11008674 DOI: 10.1016/j.mehy.2024.111322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Inflamm-aging is a condition of low-grade and chronic systemic inflammation characterized by a systemic increase in multiple inflammatory biomarkers such as tumor necrosis factor (TNF), interleukin 6 (IL-6), C-reactive protein (CRP), and CXCL9 (MIG) in experimental and clinical settings. However, despite the recent identification of extracellular procathepsin L (pCTS-L) as a novel mediator of inflammatory diseases such as sepsis, its possible role in inflamm-aging was previously not investigated. In the present study, we compared blood levels of pCTS-L and other 62 cytokines and chemokines between young and aged Balb/C mice by Western blotting and Cytokine Antibody Arrays. In light of the surprising finding of a marked increase in blood pCTS-L levels in aged mice, we propose that blood pCTS-L levels may serve as another biomarker of inflamm-aging. Given the capacity of pCTS-L in inducing various cytokines (e.g., TNF and IL-6), it will be important to test the hypothetic role of pCTS-L in inflamm-aging under experimental and clinical conditions.
Collapse
Affiliation(s)
- Cassie Shu Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Li Lou
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| |
Collapse
|
19
|
Domblides C, Crampton S, Liu H, Bartleson JM, Nguyen A, Champagne C, Landy EE, Spiker L, Proffitt C, Bhattarai S, Grawe AP, Fuentealba Valenzuela M, Lartigue L, Mahouche I, Dupaul-Chicoine J, Nishimura K, Lefort F, Decraecker M, Velasco V, Netzer S, Pitard V, Roy C, Soubeyran I, Racine V, Blanco P, Déchanet-Merville J, Saleh M, Canna SW, Furman D, Faustin B. Human NLRC4 expression promotes cancer survival and associates with type I interferon signaling and immune infiltration. J Clin Invest 2024; 134:e166085. [PMID: 38652550 PMCID: PMC11142746 DOI: 10.1172/jci166085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
The immune system can control cancer progression. However, even though some innate immune sensors of cellular stress are expressed intrinsically in epithelial cells, their potential role in cancer aggressiveness and subsequent overall survival in humans is mainly unknown. Here, we show that nucleotide-binding oligomerization domain-like receptor (NLR) family CARD domain-containing 4 (NLRC4) is downregulated in epithelial tumor cells of patients with colorectal cancer (CRC) by using spatial tissue imaging. Strikingly, only the loss of tumor NLRC4, but not stromal NLRC4, was associated with poor immune infiltration (mainly DCs and CD4+ and CD8+ T cells) and accurately predicted progression to metastatic stage IV and decrease in overall survival. By combining multiomics approaches, we show that restoring NLRC4 expression in human CRC cells triggered a broad inflammasome-independent immune reprogramming consisting of type I interferon (IFN) signaling genes and the release of chemokines and myeloid growth factors involved in the tumor infiltration and activation of DCs and T cells. Consistently, such reprogramming in cancer cells was sufficient to directly induce maturation of human DCs toward a Th1 antitumor immune response through IL-12 production in vitro. In multiple human carcinomas (colorectal, lung, and skin), we confirmed that NLRC4 expression in patient tumors was strongly associated with type I IFN genes, immune infiltrates, and high microsatellite instability. Thus, we shed light on the epithelial innate immune sensor NLRC4 as a therapeutic target to promote an efficient antitumor immune response against the aggressiveness of various carcinomas.
Collapse
Affiliation(s)
- Charlotte Domblides
- University of Bordeaux, Bordeaux, France
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
- Department of Medical Oncology, University Hospital of Bordeaux, Bordeaux, France
| | - Steven Crampton
- Discovery Immunology, Johnson & Johnson Innovative Medicine, San Diego, California, USA
| | - Hong Liu
- GI and Immune-Oncology DDUs, Takeda Pharmaceuticals, San Diego, California, and Cambridge, Massachusetts, USA
| | | | - Annie Nguyen
- Discovery Immunology, Johnson & Johnson Innovative Medicine, San Diego, California, USA
| | | | - Emily E. Landy
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lindsey Spiker
- Department of Genetics, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Christopher Proffitt
- GI and Immune-Oncology DDUs, Takeda Pharmaceuticals, San Diego, California, and Cambridge, Massachusetts, USA
| | - Sunil Bhattarai
- Discovery Immunology, Johnson & Johnson Innovative Medicine, San Diego, California, USA
| | - Anissa P. Grawe
- Buck Institute for Research on Aging, Novato, California, USA
| | | | | | | | | | - Kazuho Nishimura
- GI and Immune-Oncology DDUs, Takeda Pharmaceuticals, San Diego, California, and Cambridge, Massachusetts, USA
| | - Félix Lefort
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
- Department of Medical Oncology, University Hospital of Bordeaux, Bordeaux, France
| | - Marie Decraecker
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
| | - Valérie Velasco
- Comprehensive Cancer Center, Department of Biopathology, Institut Bergonié, Bordeaux, France
| | - Sonia Netzer
- University of Bordeaux, Bordeaux, France
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
| | - Vincent Pitard
- University of Bordeaux, Bordeaux, France
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
| | - Christian Roy
- GI and Immune-Oncology DDUs, Takeda Pharmaceuticals, San Diego, California, and Cambridge, Massachusetts, USA
| | - Isabelle Soubeyran
- Comprehensive Cancer Center, Department of Biopathology, Institut Bergonié, Bordeaux, France
| | - Victor Racine
- QuantaCell, Hôpital Saint Eloi, IRMB, Montpellier, France
| | - Patrick Blanco
- University of Bordeaux, Bordeaux, France
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
| | - Julie Déchanet-Merville
- University of Bordeaux, Bordeaux, France
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
| | - Maya Saleh
- INRS Santé Biotechnologie, Laval, Québec, Canada
| | - Scott W. Canna
- Pediatric Rheumatology, The Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, California, USA
- Stanford 1000 Immunomes Project, Stanford School of Medicine, Stanford, California, USA
| | - Benjamin Faustin
- University of Bordeaux, Bordeaux, France
- ImmunoConcEpt, CNRS UMR 5164, INSERM ERL 1303, Bordeaux University, Bordeaux, France
- Discovery Immunology, Johnson & Johnson Innovative Medicine, San Diego, California, USA
| |
Collapse
|
20
|
Sayegh S, Fantecelle CH, Laphanuwat P, Subramanian P, Rustin MHA, Gomes DCO, Akbar AN, Chambers ES. Vitamin D 3 inhibits p38 MAPK and senescence-associated inflammatory mediator secretion by senescent fibroblasts that impacts immune responses during ageing. Aging Cell 2024; 23:e14093. [PMID: 38287646 PMCID: PMC11019144 DOI: 10.1111/acel.14093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 01/31/2024] Open
Abstract
Vitamin D3 replacement in older insufficient adults significantly improves their antigen-specific varicella zoster virus (VZV) cutaneous immunity. However, the mechanisms involved in this enhancement of cutaneous immunity are not known. Here, we show for the first time that vitamin D3 blocks the senescence-associated secretory phenotype (SASP) production by senescent fibroblasts by partially inhibiting the p38 MAPK pathway. Furthermore, transcriptomic analysis of skin biopsies from older subjects after vitamin D3 supplementation shows that vitamin D3 inhibits the same inflammatory pathways in response to saline as the specific p38 inhibitor, losmapimod, which also enhances immunity in the skin of older subjects. Vitamin D3 supplementation therefore may enhance immunity during ageing in part by blocking p38 MAPK signalling and in turn inhibit SASP production from senescent cells in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel C. O. Gomes
- Núcleo de Doenças InfecciosasUniversidade Federal do Espírito SantoVitoriaBrazil
| | - Arne N. Akbar
- Division of MedicineUniversity College LondonLondonUK
| | - Emma S. Chambers
- Centre for Immunobiology, Blizard InstituteQueen Mary University of LondonLondonUK
| |
Collapse
|
21
|
Dalhat MH, Narayan S, Serio H, Arango D. Dissecting the oncogenic properties of essential RNA-modifying enzymes: a focus on NAT10. Oncogene 2024; 43:1077-1086. [PMID: 38409550 PMCID: PMC11092965 DOI: 10.1038/s41388-024-02975-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
Chemical modifications of ribonucleotides significantly alter the physicochemical properties and functions of RNA. Initially perceived as static and essential marks in ribosomal RNA (rRNA) and transfer RNA (tRNA), recent discoveries unveiled a dynamic landscape of RNA modifications in messenger RNA (mRNA) and other regulatory RNAs. These findings spurred extensive efforts to map the distribution and function of RNA modifications, aiming to elucidate their distribution and functional significance in normal cellular homeostasis and pathological states. Significant dysregulation of RNA modifications is extensively documented in cancers, accentuating the potential of RNA-modifying enzymes as therapeutic targets. However, the essential role of several RNA-modifying enzymes in normal physiological functions raises concerns about potential side effects. A notable example is N-acetyltransferase 10 (NAT10), which is responsible for acetylating cytidines in RNA. While emerging evidence positions NAT10 as an oncogenic factor and a potential target in various cancer types, its essential role in normal cellular processes complicates the development of targeted therapies. This review aims to comprehensively analyze the essential and oncogenic properties of NAT10. We discuss its crucial role in normal cell biology and aging alongside its contribution to cancer development and progression. We advocate for agnostic approaches to disentangling the intertwined essential and oncogenic functions of RNA-modifying enzymes. Such approaches are crucial for understanding the full spectrum of RNA-modifying enzymes and imperative for designing effective and safe therapeutic strategies.
Collapse
Affiliation(s)
- Mahmood H Dalhat
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Sharath Narayan
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, USA
| | - Hannah Serio
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
22
|
Kumar AAW, Huangfu G, Figtree GA, Dwivedi G. Atherosclerosis as the Damocles' sword of human evolution: insights from nonhuman ape-like primates, ancient human remains, and isolated modern human populations. Am J Physiol Heart Circ Physiol 2024; 326:H821-H831. [PMID: 38305751 DOI: 10.1152/ajpheart.00744.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
Atherosclerosis is the leading cause of death worldwide, and the predominant risk factors are advanced age and high-circulating low-density lipoprotein cholesterol (LDL-C). However, the findings of atherosclerosis in relatively young mummified remains and a lack of atherosclerosis in chimpanzees despite high LDL-C call into question the role of traditional cardiovascular risk factors. The inflammatory theory of atherosclerosis may explain the discrepancies between traditional risk factors and observed phenomena in current literature. Following the divergence from chimpanzees several millennia ago, loss of function mutations in immune regulatory genes and changes in gene expression have resulted in an overactive human immune system. The ubiquity of atherosclerosis in the modern era may reflect a selective pressure that enhanced the innate immune response at the cost of atherogenesis and other chronic disease states. Evidence provided from the fields of genetics, evolutionary biology, and paleoanthropology demonstrates a sort of circular dependency between inflammation, immune system functioning, and evolution at both a species and cellular level. More recently, the role of proinflammatory stimuli, somatic mutations, and the gene-environment effect appear to be underappreciated elements in the development and progression of atherosclerosis. Neurobiological stress, metabolic syndrome, and traditional cardiovascular risk factors may instead function as intermediary links between inflammation and atherosclerosis. Therefore, considering evolution as a mechanistic process and atherosclerosis as part of the inertia of evolution, greater insight into future preventative and therapeutic interventions for atherosclerosis can be gained by examining the past.
Collapse
Affiliation(s)
- Annora Ai-Wei Kumar
- Medical School, The University of Western Australia, Crawley, Western Australia, Australia
| | - Gavin Huangfu
- Medical School, The University of Western Australia, Crawley, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
- Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
| | - Gemma A Figtree
- Cardiovascular Discovery Group, Kolling Institute of Medical Research, St. Leonards, New South Wales, Australia
- Department of Cardiology, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| | - Girish Dwivedi
- Medical School, The University of Western Australia, Crawley, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
- Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
| |
Collapse
|
23
|
Wachter F, Pikman Y. Pathophysiology of Acute Myeloid Leukemia. Acta Haematol 2024; 147:229-246. [PMID: 38228114 DOI: 10.1159/000536152] [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: 07/25/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a biologically heterogenous disease arising in clonally proliferating hematopoietic stem cells. Sequential acquisition of mutations leads to expanded proliferation of clonal myeloid progenitors and failure of differentiation, leading to fulminant AML. SUMMARY Here, we review the pathophysiology of AML with a focus on factors predisposing to AML development, including prior chemo- and radiation therapy, environmental factors, and germline predisposition. KEY MESSAGE Increasing genomic characterization of AML and insight into mechanisms of its development will be critical to improvement in AML prognostication and therapy.
Collapse
Affiliation(s)
- Franziska Wachter
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Yana Pikman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Zhang H, Lu R, Huang J, Li L, Cao Y, Huang C, Chen R, Wang Y, Huang J, Zhao X, Yu J. N4-acetylcytidine modifies primary microRNAs for processing in cancer cells. Cell Mol Life Sci 2024; 81:73. [PMID: 38308713 PMCID: PMC10838262 DOI: 10.1007/s00018-023-05107-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/18/2023] [Accepted: 12/25/2023] [Indexed: 02/05/2024]
Abstract
N4 acetylcytidine (ac4C) modification mainly occurs on tRNA, rRNA, and mRNA, playing an important role in the expression of genetic information. However, it is still unclear whether microRNAs have undergone ac4C modification and their potential physiological and pathological functions. In this study, we identified that NAT10/THUMPD1 acetylates primary microRNAs (pri-miRNAs) with ac4C modification. Knockdown of NAT10 suppresses and augments the expression levels of mature miRNAs and pri-miRNAs, respectively. Molecular mechanism studies found that pri-miRNA ac4C promotes the processing of pri-miRNA into precursor miRNA (pre-miRNA) by enhancing the interaction of pri-miRNA and DGCR8, thereby increasing the biogenesis of mature miRNA. Knockdown of NAT10 attenuates the oncogenic characters of lung cancer cells by regulating miRNA production in cancers. Moreover, NAT10 is highly expressed in various clinical cancers and negatively correlated with poor prognosis. Thus, our results reveal that NAT10 plays a crucial role in cancer initiation and progression by modulating pri-miRNA ac4C to affect miRNA production, which would provide an attractive therapeutic strategy for cancers.
Collapse
Affiliation(s)
- Hailong Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Translational Medicine, National Center for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Runhui Lu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiayi Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lian Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingting Cao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Caihu Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ran Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanli Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xian Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
25
|
Bakker L, Choe K, Eussen SJPM, Ramakers IHGB, van den Hove DLA, Kenis G, Rutten BPF, Verhey FRJ, Köhler S. Relation of the kynurenine pathway with normal age: A systematic review. Mech Ageing Dev 2024; 217:111890. [PMID: 38056721 DOI: 10.1016/j.mad.2023.111890] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND The kynurenine pathway (KP) is gaining more attention as a common pathway involved in age-related conditions. However, which changes in the KP occur due to normal ageing is still largely unclear. The aim of this systematic review was to summarize the available evidence for associations of KP metabolites with age. METHODS We used an broad search strategy and included studies up to October 2023. RESULTS Out of 8795 hits, 55 studies were eligible for the systematic review. These studies suggest that blood levels of tryptophan decrease with age, while blood and cerebrospinal fluid levels of kynurenine and its ratio with tryptophan increase. Studies investigating associations between cerebrospinal fluid and blood levels of kynurenic acid and quinolinic acid with age reported either positive or non-significant findings. However, there is a large heterogeneity across studies. Additionally, most studies were cross-sectional, and only few studies investigated associations with other downstream kynurenines. CONCLUSIONS This systematic review suggests that levels of kynurenines are positively associated with age. Larger and prospective studies are needed that also investigate a more comprehensive panel of KP metabolites and changes during the life-course.
Collapse
Affiliation(s)
- Lieke Bakker
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands; Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands
| | - Kyonghwan Choe
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, Maastricht University, 6229 HA Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM) and Care and Public Health Research Institute (CAPHRI), 6229 ER Maastricht, the Netherlands
| | - Inez H G B Ramakers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands; Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands
| | - Daniel L A van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands; Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands; Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, 6229 ER Maastricht, the Netherlands; Alzheimer Center Limburg, Maastricht University, 6229 ET Maastricht, the Netherlands.
| |
Collapse
|
26
|
Matico RE, Yu X, Miller R, Somani S, Ricketts MD, Kumar N, Steele RA, Medley Q, Berger S, Faustin B, Sharma S. Structural basis of the human NAIP/NLRC4 inflammasome assembly and pathogen sensing. Nat Struct Mol Biol 2024; 31:82-91. [PMID: 38177670 PMCID: PMC10803261 DOI: 10.1038/s41594-023-01143-z] [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: 02/16/2023] [Accepted: 09/28/2023] [Indexed: 01/06/2024]
Abstract
The NLR family caspase activation and recruitment domain-containing 4 (NLRC4) inflammasome is a critical cytosolic innate immune machine formed upon the direct sensing of bacterial infection and in response to cell stress during sterile chronic inflammation. Despite its major role in instigating the subsequent host immune response, a more complete understanding of the molecular events in the formation of the NLRC4 inflammasome in humans is lacking. Here we identify Bacillus thailandensis type III secretion system needle protein (Needle) as a potent trigger of the human NLR family apoptosis inhibitory protein (NAIP)/NLRC4 inflammasome complex formation and determine its structural features by cryogenic electron microscopy. We also provide a detailed understanding of how type III secretion system pathogen components are sensed by human NAIP to form a cascade of NLRC4 protomer through a critical lasso-like motif, a 'lock-key' activation model and large structural rearrangement, ultimately forming the full human NLRC4 inflammasome. These results shed light on key regulatory mechanisms specific to the NLRC4 inflammasome assembly, and the innate immune modalities of pathogen sensing in humans.
Collapse
Affiliation(s)
- Rosalie E Matico
- Structural and Protein Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| | - Xiaodi Yu
- Structural and Protein Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA.
| | - Robyn Miller
- Structural and Protein Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| | - Sandeep Somani
- In Silico Discovery Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| | - M Daniel Ricketts
- Structural and Protein Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| | - Nikit Kumar
- Structural and Protein Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| | - Ruth A Steele
- Structural and Protein Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| | - Quintus Medley
- Discovery Immunology, Johnson & Johnson Innovative Medicine, Cambridge, MA, USA
| | - Scott Berger
- Discovery Immunology, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| | - Benjamin Faustin
- Discovery Immunology, Johnson & Johnson Innovative Medicine, San Diego, CA, USA
| | - Sujata Sharma
- Structural and Protein Sciences, Johnson & Johnson Innovative Medicine, Spring House, PA, USA
| |
Collapse
|
27
|
Kruglov V, Jang IH, Camell CD. Inflammaging and fatty acid oxidation in monocytes and macrophages. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00038. [PMID: 38249577 PMCID: PMC10798594 DOI: 10.1097/in9.0000000000000038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Fatty acid oxidation (FAO), primarily known as β-oxidation, plays a crucial role in breaking down fatty acids within mitochondria and peroxisomes to produce cellular energy and preventing metabolic dysfunction. Myeloid cells, including macrophages, microglia, and monocytes, rely on FAO to perform essential cellular functions and uphold tissue homeostasis. As individuals age, these cells show signs of inflammaging, a condition that includes a chronic onset of low-grade inflammation and a decline in metabolic function. These lead to changes in fatty acid metabolism and a decline in FAO pathways. Recent studies have shed light on metabolic shifts occurring in macrophages and monocytes during aging, correlating with an altered tissue environment and the onset of inflammaging. This review aims to provide insights into the connection of inflammatory pathways and altered FAO in macrophages and monocytes from older organisms. We describe a model in which there is an extended activation of receptor for advanced glycation end products, nuclear factor-κB (NF-κB) and the nod-like receptor family pyrin domain containing 3 inflammasome within macrophages and monocytes. This leads to an increased level of glycolysis, and also promotes pro-inflammatory cytokine production and signaling. As a result, FAO-related enzymes such as 5' AMP-activated protein kinase and peroxisome proliferator-activated receptor-α are reduced, adding to the escalation of inflammation, accumulation of lipids, and heightened cellular stress. We examine the existing body of literature focused on changes in FAO signaling within macrophages and monocytes and their contribution to the process of inflammaging.
Collapse
Affiliation(s)
- Victor Kruglov
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - In Hwa Jang
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Christina D. Camell
- Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
28
|
Liu M, Wei X, Xing X, Cheng Y, Ma Z, Ren J, Gao X, Xu A. Predicting fracture risk for elderly osteoporosis patients by hybrid machine learning model. Digit Health 2024; 10:20552076241257456. [PMID: 38798883 PMCID: PMC11119487 DOI: 10.1177/20552076241257456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Background and Objective Osteoporotic fractures significantly impact individuals's quality of life and exert substantial pressure on the social pension system. This study aims to develop prediction models for osteoporotic fracture and uncover potential risk factors based on Electronic Health Records (EHR). Methods Data of patients with osteoporosis were extracted from the EHR of Xinhua Hospital (July 2012-October 2017). Demographic and clinical features were used to develop prediction models based on 12 independent machine learning (ML) algorithms and 3 hybrid ML models. To facilitate a nuanced interpretation of the results, a comprehensive importance score was conceived, incorporating various perspectives to effectively discern and mine critical features from the data. Results A total of 8530 patients with osteoporosis were included for analysis, of which 1090 cases (12.8%) were fracture patients. The hybrid model that synergistically combines the Support Vector Machine (SVM) and XGBoost algorithms demonstrated the best predictive performance in terms of accuracy and precision (above 90%) among all benchmark models. Blood Calcium, Alkaline phosphatase (ALP), C-reactive Protein (CRP), Apolipoprotein A/B ratio and High-density lipoprotein cholesterol (HDL-C) were statistically found to be associated with osteoporotic fracture. Conclusions The hybrid machine learning model can be a reliable tool for predicting the risk of fracture in patients with osteoporosis. It is expected to assist clinicians in identifying high-risk fracture patients and implementing early interventions.
Collapse
Affiliation(s)
- Menghan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Wei
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaodong Xing
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yunlong Cheng
- Shanghai Key Laboratory of Scalable Computing and Systems, Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zicheng Ma
- Zhiyuan College, Shanghai Jiao Tong University, Shanghai, China
| | - Jiwu Ren
- Zhiyuan College, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofeng Gao
- Shanghai Key Laboratory of Scalable Computing and Systems, Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ajing Xu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Nassour J, Przetocka S, Karlseder J. Telomeres as hotspots for innate immunity and inflammation. DNA Repair (Amst) 2024; 133:103591. [PMID: 37951043 PMCID: PMC10842095 DOI: 10.1016/j.dnarep.2023.103591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/05/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
Aging is marked by the gradual accumulation of deleterious changes that disrupt organ function, creating an altered physiological state that is permissive for the onset of prevalent human diseases. While the exact mechanisms governing aging remain a subject of ongoing research, there are several cellular and molecular hallmarks that contribute to this biological process. This review focuses on two factors, namely telomere dysfunction and inflammation, which have emerged as crucial contributors to the aging process. We aim to discuss the mechanistic connections between these two distinct hallmarks and provide compelling evidence highlighting the loss of telomere protection as a driver of pro-inflammatory states associated with aging. By reevaluating the interplay between telomeres, innate immunity, and inflammation, we present novel perspectives on the etiology of aging and its associated diseases.
Collapse
Affiliation(s)
- Joe Nassour
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, 12801 E. 17th Ave, Aurora, CO 80045, USA; The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Sara Przetocka
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Jan Karlseder
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA.
| |
Collapse
|
30
|
Connors J, Cusimano G, Mege N, Woloszczuk K, Konopka E, Bell M, Joyner D, Marcy J, Tardif V, Kutzler MA, Muir R, Haddad EK. Using the power of innate immunoprofiling to understand vaccine design, infection, and immunity. Hum Vaccin Immunother 2023; 19:2267295. [PMID: 37885158 PMCID: PMC10760375 DOI: 10.1080/21645515.2023.2267295] [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: 07/13/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
In the field of immunology, a systems biology approach is crucial to understanding the immune response to infection and vaccination considering the complex interplay between genetic, epigenetic, and environmental factors. Significant progress has been made in understanding the innate immune response, including cell players and critical signaling pathways, but many questions remain unanswered, including how the innate immune response dictates host/pathogen responses and responses to vaccines. To complicate things further, it is becoming increasingly clear that the innate immune response is not a linear pathway but is formed from complex networks and interactions. To further our understanding of the crosstalk and complexities, systems-level analyses and expanded experimental technologies are now needed. In this review, we discuss the most recent immunoprofiling techniques and discuss systems approaches to studying the global innate immune landscape which will inform on the development of personalized medicine and innovative vaccine strategies.
Collapse
Affiliation(s)
- Jennifer Connors
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Gina Cusimano
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Nathan Mege
- Tower Health, Reading Hospital, West Reading, PA, USA
| | - Kyra Woloszczuk
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Emily Konopka
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Matthew Bell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - David Joyner
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Molecular and Cellular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jennifer Marcy
- Department of Molecular and Cellular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Virginie Tardif
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michele A. Kutzler
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Roshell Muir
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Family, Community, and Preventative Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Elias K. Haddad
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
31
|
Zhang B, Wan H, Liu X, Yu T, Yang Y, Dai Y, Han Y, Xu K, Yang L, Wang Y, Zhang X. Engineering Immunomodulatory Stents Using Zinc Ion-Lysozyme Nanoparticle Platform for Vascular Remodeling. ACS NANO 2023; 17:23498-23511. [PMID: 37971533 DOI: 10.1021/acsnano.3c06103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Rapid endothelialization of cardiovascular materials can enhance the vascular remodeling performance. In this work, we developed a strategy for amyloid-like protein-assembly-mediated interfacial engineering to functionalize a biomimetic nanoparticle coating (BMC). Various groups (e.g., hydroxyl and carboxyl) on the BMC are responsible for chelating Zn2+ ions at the stent interface, similar to the glutathione peroxidase-like enzymes found in vivo. This design could reproduce the release of therapeutic nitric oxide gas (NO) and an aligned microenvironment nearly identical with that of natural vessels. In a rabbit abdominal aorta model, BMC-coated stents promoted vascular healing through rapid endothelialization and the inhibition of intimal hyperplasia in the placement sites at 4, 12, and 24 weeks. Additionally, better anticoagulant activity and immunomodulation in the BMC stents were also confirmed, and vascular healing was mainly dependent on cell signaling through the cyclic guanosine monophosphate-protein kinase G (cGMP-PKG) cascade. Overall, a metal-polypeptide-coated stent was developed on the basis of its detailed molecular mechanism of action in vascular remodeling.
Collapse
Affiliation(s)
- Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Huining Wan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Xiyu Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Tao Yu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Yuan Yang
- Sichuan Xingtai Pule Medical Technology Co Ltd, Chengdu, Sichuan 610045, China
| | - Yan Dai
- Sichuan Xingtai Pule Medical Technology Co Ltd, Chengdu, Sichuan 610045, China
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang 110016, China
| | - Kai Xu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Wenhua Road 83, Shenyang 110016, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| |
Collapse
|
32
|
Zhong X, Qiang Y, Wang L, Zhang Y, Li J, Feng J, Cheng W, Tan L, Yu J. Peripheral immunity and risk of incident brain disorders: a prospective cohort study of 161,968 participants. Transl Psychiatry 2023; 13:382. [PMID: 38071240 PMCID: PMC10710500 DOI: 10.1038/s41398-023-02683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Whether peripheral immunity prospectively influences brain health remains controversial. This study aims to investigate the longitudinal associations between peripheral immunity markers with incident brain disorders. A total of 161,968 eligible participants from the UK Biobank were included. We investigated the linear and non-linear effects of peripheral immunity markers including differential leukocytes counts, their derived ratios and C-reactive protein (CRP) on the risk of dementia, Parkinson's disease (PD), stroke, schizophrenia, bipolar affective disorder (BPAD), major depressive disorder (MDD) and anxiety, using Cox proportional hazard models and restricted cubic spline models. Linear regression models were used to explore potential mechanisms driven by brain structures. During a median follow-up of 9.66 years, 16,241 participants developed brain disorders. Individuals with elevated innate immunity markers including neutrophils, monocytes, platelets, neutrophil-to-lymphocyte ratio (NLR), and systemic immune-inflammation index (SII) had an increased risk of brain disorders. Among these markers, neutrophils exhibited the most significant correlation with risk of dementia (hazard ratio 1.08, 95% confidence interval 1.04-1.12), stroke (HR 1.06, 95% CI 1.03-1.09), MDD (HR 1.13, 95% CI 1.10-1.16) and anxiety (HR 1.07, 95% CI 1.04-1.10). Subgroup analysis revealed age-specific and sex-specific associations between innate immunity markers with risk of dementia and MDD. Neuroimaging analysis highlighted the associations between peripheral immunity markers and alterations in multiple cortical, subcortical regions and white matter tracts, typically implicated in dementia and psychiatric disorders. These findings support the hypothesis that neuroinflammation is important to the etiology of various brain disorders, offering new insights into their potential therapeutic approaches.
Collapse
Affiliation(s)
- Xiaoling Zhong
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, China
| | - Yixuan Qiang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological diseases, Shanghai, China
| | - Ling Wang
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, China
| | - Yaru Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological diseases, Shanghai, China
| | - Jieqiong Li
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jianfeng Feng
- The Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Wei Cheng
- The Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Jintai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological diseases, Shanghai, China.
| |
Collapse
|
33
|
Shi J, Yang C, Zhang J, Zhao K, Li P, Kong C, Wu X, Sun H, Zheng R, Sun W, Chen L, Kong X. NAT10 Is Involved in Cardiac Remodeling Through ac4C-Mediated Transcriptomic Regulation. Circ Res 2023; 133:989-1002. [PMID: 37955115 DOI: 10.1161/circresaha.122.322244] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 10/30/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Heart failure, characterized by cardiac remodeling, is associated with abnormal epigenetic processes and aberrant gene expression. Here, we aimed to elucidate the effects and mechanisms of NAT10 (N-acetyltransferase 10)-mediated N4-acetylcytidine (ac4C) acetylation during cardiac remodeling. METHODS NAT10 and ac4C expression were detected in both human and mouse subjects with cardiac remodeling through multiple assays. Subsequently, acetylated RNA immunoprecipitation and sequencing, thiol-linked alkylation for the metabolic sequencing of RNA (SLAM-seq), and ribosome sequencing (Ribo-seq) were employed to elucidate the role of ac4C-modified posttranscriptional regulation in cardiac remodeling. Additionally, functional experiments involving the overexpression or knockdown of NAT10 were conducted in mice models challenged with Ang II (angiotensin II) and transverse aortic constriction. RESULTS NAT10 expression and RNA ac4C levels were increased in in vitro and in vivo cardiac remodeling models, as well as in patients with cardiac hypertrophy. Silencing and inhibiting NAT10 attenuated Ang II-induced cardiomyocyte hypertrophy and cardiofibroblast activation. Next-generation sequencing revealed ac4C changes in both mice and humans with cardiac hypertrophy were associated with changes in global mRNA abundance, stability, and translation efficiency. Mechanistically, NAT10 could enhance the stability and translation efficiency of CD47 and ROCK2 transcripts by upregulating their mRNA ac4C modification, thereby resulting in an increase in their protein expression during cardiac remodeling. Furthermore, the administration of Remodelin, a NAT10 inhibitor, has been shown to prevent cardiac functional impairments in mice subjected to transverse aortic constriction by suppressing cardiac fibrosis, hypertrophy, and inflammatory responses, while also regulating the expression levels of CD47 and ROCK2 (Rho associated coiled-coil containing protein kinase 2). CONCLUSIONS Therefore, our data suggest that modulating epitranscriptomic processes, such as ac4C acetylation through NAT10, may be a promising therapeutic target against cardiac remodeling.
Collapse
Affiliation(s)
- Jing Shi
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Chuanxi Yang
- Department of Cardiology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China (C.Y.)
| | - Jing Zhang
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Kun Zhao
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Peng Li
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Chuiyu Kong
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Jiangsu, China (C.K.)
| | - Xiaoguang Wu
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Haoliang Sun
- Department of Cardiovascular Surgery (H.S., R.Z.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Rui Zheng
- Department of Cardiovascular Surgery (H.S., R.Z.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Wei Sun
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
| | - Lianmin Chen
- Changzhou Medical Center of the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University and Department of Cardiology of the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China (L.C.)
| | - Xiangqing Kong
- Department of Cardiology (J.S., K.Z., J.Z., P.L., X.W., W.S., X.K.), The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, China
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China (X.K.)
| |
Collapse
|
34
|
Morelli V, Heizelman RJ. Monitoring Social Determinants of Health Assessing Patients and Communities. Prim Care 2023; 50:527-547. [PMID: 37866829 DOI: 10.1016/j.pop.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Because of the devastating health effects of social determinants of health (SDoH), it is important for the primary care provider to assess and monitor these types of stressors. This can be done via surveys, geomapping, or various biomarkers. To date, however, each of these methods is fraught with obstacles. There are currently are no validated "best" SDoH screening tools for use in clinical practice. Nor is geomapping, a perfect solution. Although mapping can collect location specific factors, it does not account for the fact that patients may live in one area, work in another and travel frequently to a third.
Collapse
Affiliation(s)
- Vincent Morelli
- Department of Family and Community Medicine, Meharry Medical College, 3rd Floor, Old Hospital Building, 1005 Dr. D. B. Todd, Jr., Boulevard, Nashville, TN 37208-3599, USA.
| | - Robert Joseph Heizelman
- Department of Family Medicine, Medical Informatics, University of Michigan, 3rd Floor, Old Hospital Building, 1005 Dr. D. B. Todd, Jr., Boulevard, Nashville, TN 37208-3599, USA
| |
Collapse
|
35
|
Gholami A, Amirkalali B, Baradaran HR, Hariri M. A systematic review and dose-response meta-analysis of the effect of selenium supplementation on serum concentration of C-reactive protein. J Trace Elem Med Biol 2023; 80:127273. [PMID: 37531798 DOI: 10.1016/j.jtemb.2023.127273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 06/12/2023] [Accepted: 07/23/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Despite conducting new studies on the potential effect of selenium in reducing inflammation; results held contradictory weights and turns into an intriguing area of research. Concerns have been raised on this matter on hand to reach conclusive/consistent results. Therefore, this study seeks to address this scientific gap by assessing randomized controlled trials (RCTs) investigating the effect of selenium intake on C-reactive protein (CRP). METHOD AND MATERIALS To find all the relevant English-language RCTs, databases including SCOPUS, Web of Science, Clinical Trials.gov, PubMed, and Cochrane Library were searched with no time limit (up to June 2022). Based on the mean changes for both comparison and intervention groups, the effect sizes were calculated. Further, DerSimonian and Laird random-effects model was performed to determine the summary of overall effects and their heterogeneity. RESULTS Fifteen studies were selected for systematic review and thirteen studies were known to be eligible for meta-analysis. Meta-analysis results indicated a significant effect in serum CRP concentrations compared to the control group (weighted mean difference (WMD)= -0.22 mg/L 95 % confidence interval (CI): - 0.39, - 0.04; p = 0.014). CONCLUSION In conclusion, our study confirms the downward effect of selenium on the serum concentration of CRP. On a wider level, further research is also needed to assess the selenium effect on other inflammatory mediators.
Collapse
Affiliation(s)
- Ali Gholami
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran; Department of Epidemiology and Biostatistics, School of Public Health, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Bahareh Amirkalali
- Gastrointestinal & Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Baradaran
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Ageing Clinical and Experimental Research Team, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen, Aberdeen, UK
| | - Mitra Hariri
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran; Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
36
|
Hu Y, Liu Y, Zheng H, Liu L. Risk Factors for Long COVID in Older Adults. Biomedicines 2023; 11:3002. [PMID: 38002002 PMCID: PMC10669899 DOI: 10.3390/biomedicines11113002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
As time has passed following the COVID-19 pandemic, individuals infected with SARS-CoV-2 have gradually exhibited a variety of symptoms associated with long COVID in the postacute phase of infection. Simultaneously, in many countries worldwide, the process of population aging has been accelerating. Within this context, the elderly population has not only become susceptible and high-risk during the acute phase of COVID-19 but also has considerable risks when confronting long COVID. Elderly individuals possess specific immunological backgrounds, and during the process of aging, their immune systems can enter a state known as "immunosenescence". This further exacerbates "inflammaging" and the development of various comorbidities in elderly individuals, rendering them more susceptible to long COVID. Additionally, long COVID can inflict both physical and mental harm upon elderly people, thereby reducing their overall quality of life. Consequently, the impact of long COVID on elderly people should not be underestimated. This review seeks to summarize the infection characteristics and intrinsic factors of older adults during the COVID-19 pandemic, with a focus on the physical and mental impact of long COVID. Additionally, it aims to explore potential strategies to mitigate the risk of long COVID or other emerging infectious diseases among older adults in the future.
Collapse
Affiliation(s)
| | | | | | - Longding Liu
- Key Laboratory of Systemic Innovative Research on Virus Vaccines, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (Y.H.); (Y.L.); (H.Z.)
| |
Collapse
|
37
|
Wan T, Wang Y, He K, Zhu S. Microbial sensing in the intestine. Protein Cell 2023; 14:824-860. [PMID: 37191444 PMCID: PMC10636641 DOI: 10.1093/procel/pwad028] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
The gut microbiota plays a key role in host health and disease, particularly through their interactions with the immune system. Intestinal homeostasis is dependent on the symbiotic relationships between the host and the diverse gut microbiota, which is influenced by the highly co-evolved immune-microbiota interactions. The first step of the interaction between the host and the gut microbiota is the sensing of the gut microbes by the host immune system. In this review, we describe the cells of the host immune system and the proteins that sense the components and metabolites of the gut microbes. We further highlight the essential roles of pattern recognition receptors (PRRs), the G protein-coupled receptors (GPCRs), aryl hydrocarbon receptor (AHR) and the nuclear receptors expressed in the intestinal epithelial cells (IECs) and the intestine-resident immune cells. We also discuss the mechanisms by which the disruption of microbial sensing because of genetic or environmental factors causes human diseases such as the inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Tingting Wan
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Yalong Wang
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Kaixin He
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Shu Zhu
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
- Department of Digestive Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| |
Collapse
|
38
|
Omer MM, Alam M, Rawat A, Lakhdhir F, Alhneif M, Rabadia D, Wei CR, Hirani S. Meta-Analysis on the Impact of Inflammatory Rheumatological Conditions on Outcomes Following Acute Coronary Syndrome. Cureus 2023; 15:e49376. [PMID: 38146576 PMCID: PMC10749408 DOI: 10.7759/cureus.49376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2023] [Indexed: 12/27/2023] Open
Abstract
Inflammatory rheumatological conditions, also known as inflammatory rheumatic conditions (IRC), constitute a category of autoimmune and inflammatory ailments primarily affecting the musculoskeletal system, encompassing the joints, muscles, and connective tissues. The objective of this meta-analysis is to evaluate the impact of inflammatory rheumatological conditions (IRC) on post-acute coronary syndrome (ACS) outcomes. This study was performed as per the preferred reporting items for systematic review and meta-analysis (PRISMA) guidelines. The PubMed, Web of Science, and Scopus databases were searched by two authors without any language constraints from January 1, 2015, to October 15, 2023. The primary outcome assessed in this meta-analysis was all-cause mortality. Other outcomes included myocardial infarction and revascularization. A total of 11 studies were included in this meta-analysis. The risk of all-cause mortality was significantly higher in patients with IRC compared to non-IRC patients (RR: 1.12, 95% CI: 1.00 to 1.26, p-value: 0.04). There is a significantly higher risk of myocardial infarction and revascularization in patients with IRC as opposed to those without IRC. Furthermore, while there was a higher risk of stroke in the IRC group compared to the non-IRC group, this disparity did not reach statistical significance. Future research should focus on specific inflammatory rheumatoid conditions, a comprehensive evaluation of cardiovascular events, and targeted interventions to enhance patient outcomes in this vulnerable population.
Collapse
Affiliation(s)
| | - Morshed Alam
- Internal Medicine, Chittagong Medical College, Chittagong, BGD
| | - Anurag Rawat
- Interventional Cardiology, Himalayan Institute of Medical Sciences, Dehradun, IND
| | - Fahad Lakhdhir
- Adult Cardiology, National Institute of Cardiovascular Diseases, Karachi, PAK
| | | | - Dhaval Rabadia
- Medicine, Surat Municipal Institute of Medical Education and Research, Surat, IND
| | - Calvin R Wei
- Research and Development, Shing Huei Group, Taipei, TWN
| | | |
Collapse
|
39
|
Zane F, Bouzid H, Sosa Marmol S, Brazane M, Besse S, Molina JL, Cansell C, Aprahamian F, Durand S, Ayache J, Antoniewski C, Todd N, Carré C, Rera M. Smurfness-based two-phase model of ageing helps deconvolve the ageing transcriptional signature. Aging Cell 2023; 22:e13946. [PMID: 37822253 PMCID: PMC10652310 DOI: 10.1111/acel.13946] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 10/13/2023] Open
Abstract
Ageing is characterised at the molecular level by six transcriptional 'hallmarks of ageing', that are commonly described as progressively affected as time passes. By contrast, the 'Smurf' assay separates high-and-constant-mortality risk individuals from healthy, zero-mortality risk individuals, based on increased intestinal permeability. Performing whole body total RNA sequencing, we found that Smurfness distinguishes transcriptional changes associated with chronological age from those associated with biological age. We show that transcriptional heterogeneity increases with chronological age in non-Smurf individuals preceding the other five hallmarks of ageing that are specifically associated with the Smurf state. Using this approach, we also devise targeted pro-longevity genetic interventions delaying entry in the Smurf state. We anticipate that increased attention to the evolutionary conserved Smurf phenotype will bring about significant advances in our understanding of the mechanisms of ageing.
Collapse
Affiliation(s)
- Flaminia Zane
- Université Paris Cité, INSERM UMR U1284ParisFrance
- Institut de Biologie Paris Seine, Sorbonne UniversitéParisFrance
| | - Hayet Bouzid
- Université Paris Cité, INSERM UMR U1284ParisFrance
- Institut de Biologie Paris Seine, Sorbonne UniversitéParisFrance
| | | | - Mira Brazane
- Institut de Biologie Paris Seine, Sorbonne UniversitéParisFrance
| | | | | | - Céline Cansell
- Université Paris‐Saclay, AgroParisTech, INRAE, UMR PNCAPalaiseauFrance
| | - Fanny Aprahamian
- Metabolomics and Cell Biology Platforms, UMS AMMICaInstitut Gustave RoussyVillejuifFrance
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le CancerUniversité de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de FranceParisFrance
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, UMS AMMICaInstitut Gustave RoussyVillejuifFrance
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le CancerUniversité de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de FranceParisFrance
| | - Jessica Ayache
- Institut Jacques Monod, CNRS UMR 7592, Université Paris CitéParisFrance
| | | | - Nicolas Todd
- Eco‐Anthropologie (EA), Muséum National d'Histoire Naturelle, CNRSUniversité de Paris, Musée de l'HommeParisFrance
| | - Clément Carré
- Institut de Biologie Paris Seine, Sorbonne UniversitéParisFrance
| | - Michael Rera
- Université Paris Cité, INSERM UMR U1284ParisFrance
| |
Collapse
|
40
|
Martyshkina YS, Tereshchenko VP, Bogdanova DA, Rybtsov SA. Reliable Hallmarks and Biomarkers of Senescent Lymphocytes. Int J Mol Sci 2023; 24:15653. [PMID: 37958640 PMCID: PMC10647376 DOI: 10.3390/ijms242115653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
The phenomenon of accumulation of senescent adaptive immunity cells in the elderly is attracting attention due to the increasing risk of global epidemics and aging of the global population. Elderly people are predisposed to various infectious and age-related diseases and are at higher risk of vaccination failure. The accumulation of senescent cells increases age-related background inflammation, "Inflammaging", causing lymphocyte exhaustion and cardiovascular, neurodegenerative, autoimmune and cancer diseases. Here, we present a comprehensive contemporary review of the mechanisms and phenotype of senescence in the adaptive immune system. Although modern research has not yet identified specific markers of aging lymphocytes, several sets of markers facilitate the separation of the aging population based on normal memory and exhausted cells for further genetic and functional analysis. The reasons for the higher predisposition of CD8+ T-lymphocytes to senescence compared to the CD4+ population are also discussed. We point out approaches for senescent-lymphocyte-targeting markers using small molecules (senolytics), antibodies and immunization against senescent cells. The suppression of immune senescence is the most relevant area of research aimed at developing anti-aging and anti-cancer therapy for prolonging the lifespan of the global population.
Collapse
Affiliation(s)
- Yuliya S. Martyshkina
- Division of Immunobiology and Biomedicine, Center for Genetics and Life Sciences, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia; (Y.S.M.)
| | - Valeriy P. Tereshchenko
- Resource Center for Cell Technology and Immunology, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia
| | - Daria A. Bogdanova
- Division of Immunobiology and Biomedicine, Center for Genetics and Life Sciences, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia; (Y.S.M.)
| | - Stanislav A. Rybtsov
- Resource Center for Cell Technology and Immunology, Sirius University of Science and Technology, Olimpiyskiy Ave. b.1, Sirius 354340, Krasnodar Region, Russia
| |
Collapse
|
41
|
Galizzi G, Di Carlo M. Mitochondrial DNA and Inflammation in Alzheimer's Disease. Curr Issues Mol Biol 2023; 45:8586-8606. [PMID: 37998717 PMCID: PMC10670154 DOI: 10.3390/cimb45110540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/15/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023] Open
Abstract
Mitochondrial dysfunction and neuroinflammation are implicated in the pathogenesis of most neurodegenerative diseases, such as Alzheimer's disease (AD). In fact, although a growing number of studies show crosstalk between these two processes, there remain numerous gaps in our knowledge of the mechanisms involved, which requires further clarification. On the one hand, mitochondrial dysfunction may lead to the release of mitochondrial damage-associated molecular patterns (mtDAMPs) which are recognized by microglial immune receptors and contribute to neuroinflammation progression. On the other hand, inflammatory molecules released by glial cells can influence and regulate mitochondrial function. A deeper understanding of these mechanisms may help identify biomarkers and molecular targets useful for the treatment of neurodegenerative diseases. This review of works published in recent years is focused on the description of the mitochondrial contribution to neuroinflammation and neurodegeneration, with particular attention to mitochondrial DNA (mtDNA) and AD.
Collapse
Affiliation(s)
- Giacoma Galizzi
- Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Via Ugo La Malfa, 153-90146 Palermo, Italy;
| | | |
Collapse
|
42
|
Maurmann RM, Schmitt BL, Mosalmanzadeh N, Pence BD. Mitochondrial dysfunction at the cornerstone of inflammatory exacerbation in aged macrophages. EXPLORATION OF IMMUNOLOGY 2023; 3:442-452. [PMID: 38831878 PMCID: PMC11147369 DOI: 10.37349/ei.2023.00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/18/2023] [Indexed: 06/05/2024]
Abstract
Immunosenescence encompasses multiple age-related adaptations that result in increased susceptibility to infections, chronic inflammatory disorders, and higher mortality risk. Macrophages are key innate cells implicated in inflammatory responses and tissue homeostasis, functions progressively compromised by aging. This process coincides with declining mitochondrial physiology, whose integrity is required to sustain and orchestrate immune responses. Indeed, multiple insults observed in aged macrophages have been implied as drivers of mitochondrial dysfunction, but how this translates into impaired immune function remains sparsely explored. This review provides a perspective on recent studies elucidating the underlying mechanisms linking dysregulated mitochondria homeostasis to immune function in aged macrophages. Genomic stress alongside defective mitochondrial turnover accounted for the progressive accumulation of damaged mitochondria in aged macrophages, thus resulting in a higher susceptibility to excessive mitochondrial DNA (mtDNA) leakage and reactive oxygen species (ROS) production. Increased levels of these mitochondrial products following infection were demonstrated to contribute to exacerbated inflammatory responses mediated by overstimulation of NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome and cyclic GMP-ATP synthase (cGAS)-stimulator of interferon genes (STING) pathways. While these mechanisms are not fully elucidated, the present evidence provides a promising area to be explored and a renewed perspective of potential therapeutic targets for immunological dysfunction.
Collapse
Affiliation(s)
| | | | - Negin Mosalmanzadeh
- College of Health Sciences, University of Memphis, Memphis, Tennessee, 38152, USA
| | - Brandt D. Pence
- College of Health Sciences, University of Memphis, Memphis, Tennessee, 38152, USA
| |
Collapse
|
43
|
Yan Q, Zhou J, Wang Z, Ding X, Ma X, Li W, Jia X, Gao SJ, Lu C. NAT10-dependent N 4-acetylcytidine modification mediates PAN RNA stability, KSHV reactivation, and IFI16-related inflammasome activation. Nat Commun 2023; 14:6327. [PMID: 37816771 PMCID: PMC10564894 DOI: 10.1038/s41467-023-42135-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
N-acetyltransferase 10 (NAT10) is an N4-acetylcytidine (ac4C) writer that catalyzes RNA acetylation at cytidine N4 position on tRNAs, rRNAs and mRNAs. Recently, NAT10 and the associated ac4C have been reported to increase the stability of HIV-1 transcripts. Here, we show that NAT10 catalyzes ac4C addition to the polyadenylated nuclear RNA (PAN), a long non-coding RNA encoded by the oncogenic DNA virus Kaposi's sarcoma-associated herpesvirus (KSHV), triggering viral lytic reactivation from latency. Mutagenesis of ac4C sites in PAN RNA in the context of KSHV infection abolishes PAN ac4C modifications, downregulates the expression of viral lytic genes and reduces virion production. NAT10 knockdown or mutagenesis erases ac4C modifications of PAN RNA and increases its instability, and prevents KSHV reactivation. Furthermore, PAN ac4C modification promotes NAT10 recruitment of IFN-γ-inducible protein-16 (IFI16) mRNA, resulting in its ac4C acetylation, mRNA stability and translation, and eventual inflammasome activation. These results reveal a novel mechanism of viral and host ac4C modifications and the associated complexes as a critical switch of KSHV replication and antiviral immunity.
Collapse
Affiliation(s)
- Qin Yan
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
- Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Jing Zhou
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Ziyu Wang
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Xiangya Ding
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China
| | - Xinyue Ma
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Wan Li
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
- Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China.
| | - Shou-Jiang Gao
- Tumor Virology Program, UPMC Hillman Cancer Center, and Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Chun Lu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China.
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China.
- Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, P. R. China.
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, 211166, Nanjing, P. R. China.
| |
Collapse
|
44
|
Wang F, Tessier AJ, Liang L, Wittenbecher C, Haslam DE, Fernández-Duval G, Heather Eliassen A, Rexrode KM, Tobias DK, Li J, Zeleznik O, Grodstein F, Martínez-González MA, Salas-Salvadó J, Clish C, Lee KH, Sun Q, Stampfer MJ, Hu FB, Guasch-Ferré M. Plasma metabolomic profiles associated with mortality and longevity in a prospective analysis of 13,512 individuals. Nat Commun 2023; 14:5744. [PMID: 37717037 PMCID: PMC10505179 DOI: 10.1038/s41467-023-41515-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 09/01/2023] [Indexed: 09/18/2023] Open
Abstract
Experimental studies reported biochemical actions underpinning aging processes and mortality, but the relevant metabolic alterations in humans are not well understood. Here we examine the associations of 243 plasma metabolites with mortality and longevity (attaining age 85 years) in 11,634 US (median follow-up of 22.6 years, with 4288 deaths) and 1878 Spanish participants (median follow-up of 14.5 years, with 525 deaths). We find that, higher levels of N2,N2-dimethylguanosine, pseudouridine, N4-acetylcytidine, 4-acetamidobutanoic acid, N1-acetylspermidine, and lipids with fewer double bonds are associated with increased risk of all-cause mortality and reduced odds of longevity; whereas L-serine and lipids with more double bonds are associated with lower mortality risk and a higher likelihood of longevity. We further develop a multi-metabolite profile score that is associated with higher mortality risk. Our findings suggest that differences in levels of nucleosides, amino acids, and several lipid subclasses can predict mortality. The underlying mechanisms remain to be determined.
Collapse
Affiliation(s)
- Fenglei Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anne-Julie Tessier
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Clemens Wittenbecher
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- SciLifeLab, Division of Food Science and Nutrition, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Danielle E Haslam
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gonzalo Fernández-Duval
- Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA), University of Navarra, Pamplona, Spain
| | - A Heather Eliassen
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathryn M Rexrode
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Deirdre K Tobias
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Oana Zeleznik
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Miguel A Martínez-González
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA), University of Navarra, Pamplona, Spain
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jordi Salas-Salvadó
- Consorcio CIBER, Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Clary Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kyu Ha Lee
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Meir J Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
45
|
Liu N, Xia L. Investigation of epigenetics insights of hypertension: A bibliometric analysis. Medicine (Baltimore) 2023; 102:e35125. [PMID: 37682151 PMCID: PMC10489307 DOI: 10.1097/md.0000000000035125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
PURPOSE Hypertension remains a major risk factor for myocardial infarction, heart failure, end-stage renal disease, and stroke. Multiple genes are involved in the process of hypertension with an additional dimension of interaction with the environment. This study conducted a bibliometric analysis of publications in the field of hypertension and epigenetics over the past 10 years to summarize the current status of the field and analyze the trends in the field. METHODS On February 5, 2023, we chose the web of science core collection database as the study data source. VOS viewer 1.6.18 and Cite Space 6.1.6 were used to examine publications of research on hypertension and epigenetics that were published between 2013 and 2022. We looked through the papers for journals, organizations, nations and regions, authors, and key terms. RESULTS This analysis covered a total of 1535 papers on studies into hypertension and epigenetics. There were 7279 authors, 83 nations, 1983 organizations, and 606 journals in all of the articles. In the USA, 540 publications were the most. The institution with the most publications was Harvard Medical School. The author with the most articles was Zhao Lubo. CONCLUSION This study summarizes the global research trends in hypertension and epigenetics. Publications in this field have increased year by year in the last decade and the field of research on hypertension and epigenetics has good prospects for growth.
Collapse
Affiliation(s)
- Nannan Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Lina Xia
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
46
|
Song Y, You Q, Chen X. Transition Metal-Based Therapies for Inflammatory Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2212102. [PMID: 36863722 DOI: 10.1002/adma.202212102] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/15/2023] [Indexed: 08/04/2023]
Abstract
Inflammatory disease (ID) is a general term that covers all diseases in which chronic inflammation performs as the major manifestation of pathogenesis. Traditional therapies based on the anti-inflammatory and immunosuppressive drugs are palliative with the short-term remission. The emergence of nanodrugs has been reported to solve the potential causes and prevent recurrences, thus holding great potential for the treatment of IDs. Among various nanomaterial systems, transition metal-based smart nanosystems (TMSNs) with unique electronic structures possess therapeutic advantages owing to their large surface area to volume ratio, high photothermal conversion efficiency, X-ray absorption capacity, and multiple catalytic enzyme activities. In this review, the rationale, design principle, and therapeutic mechanisms of TMSNs for treatments of various IDs are summarized. Specifically, TMSNs can not only be designed to scavenge danger signals, such as reactive oxygen and nitrogen species and cell-free DNA, but also can be engineered to block the mechanism of initiating inflammatory responses. In addition, TMSNs can be further applied as nanocarriers to deliver anti-inflammatory drugs. Finally, the opportunities and challenges of TMSNs are discussed, and the future directions of TMSN-based ID treatment for clinical applications are emphasized.
Collapse
Affiliation(s)
- Yilin Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Qing You
- Departments of Diagnostic, Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program NUS center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic, Radiology Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program NUS center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
47
|
Jiang M, Wang K, Huang Y, Zhang X, Yang T, Zhan K, Zhao G. Quercetin Alleviates Lipopolysaccharide-Induced Cell Oxidative Stress and Inflammatory Responses via Regulation of the TLR4-NF-κB Signaling Pathway in Bovine Rumen Epithelial Cells. Toxins (Basel) 2023; 15:512. [PMID: 37624269 PMCID: PMC10467142 DOI: 10.3390/toxins15080512] [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: 08/04/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Subacute rumen acidosis (SARA) will cause an increase in endotoxin, which will have a negative effect on the bovine rumen epithelial cells (BREC). Flavonoids are effective in treating inflammation caused by endotoxin. Quercetin is a vital flavonoid widely occurring in fruits and vegetables and has received significant interest as a prospective anti-inflammatory antioxidant. Nonetheless, quercetin's protective machinery against such damage to BREC induced by lipopolysaccharide (LPS) remains unclear. A combined quercetin and LPS-induced BREC inflammation model was utilized to elucidate the effect of quercetin protecting BREC from LPS-induced injury. After treating BREC with different doses of LPS (1, 5, and 10 μg/mL) for 6 h or 24 h, the mRNA expression of inflammatory factors was detected. Our experimental results show the establishment of the BREC inflammation model via mRNA high expression of pro-inflammatory cytokines in BREC following 6 h treatment with 1 µg/mL LPS. The promotive effect of 80 μg/mL quercetin on BREC growth via the cell counting kit-8 (CCK8) assay was observed. The expression of pro-inflammatory cytokines and chemokines, notably tumor necrosis factor α (TNF-α), Interleukin 1β (IL-1β), IL-6, CC-motif chemokine ligand 2 (CCL2), CCL20, CCL28, and CXC motif chemokine 9 (CXCL9), etc., was significantly reduced by quercetin supplementation. We also analyzed the mRNA detection of related pathways by qRT-PCR. Our validation studies demonstrated that quercetin markedly curbed the mRNA expression of the toll-like receptor 4 (TLR4) and myeloid differentiation primary response protein (MyD88) and the nuclear factor-κB (NF-κB) in LPS-treated BREC. In addition, western blot result outcomes confirmed, as expected, that LPS significantly activated phosphorylation of p44/42 extracellular regulated protein kinases (ERK1/2) and NF-κB. Unexpectedly, this effect was reversed by adding quercetin. To complement western blot results, we assessed p-ERK1/2 and p-p65 protein expression using immunofluorescence, which gave consistent results. Therefore, quercetin's capacity to bar the TLR4-mediated NF-κB and MAPK signaling pathways may be the cause of its anti-inflammatory effects on LPS-induced inflammatory reactions in BREC. According to these results, quercetin may be utilized as an anti-inflammatory medication to alleviate inflammation brought on by high-grain feed, and it also lays out a conceptual foundation regarding the development and utilization of quercetin in the later stage.
Collapse
Affiliation(s)
- Maocheng Jiang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Kexin Wang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yinghao Huang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xuelei Zhang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Tianyu Yang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Kang Zhan
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Guoqi Zhao
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
48
|
Preetam S, Jonnalagadda S, Kumar L, Rath R, Chattopadhyay S, Alghamdi BS, Abuzenadah AM, Jha NK, Gautam A, Malik S, Ashraf GM. Therapeutic potential of lipid nanosystems for the treatment of Parkinson's disease. Ageing Res Rev 2023; 89:101965. [PMID: 37268112 DOI: 10.1016/j.arr.2023.101965] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/28/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. The degeneration of dopaminergic neurons in the midbrain is primarily responsible for the onset of the disease. The major challenge faced in the treatment of PD is the blood-brain barrier (BBB), which impedes the delivery of therapeutics to targeted locations. To address this issue, lipid nanosystems have been used for the precise delivery of therapeutic compounds in anti-PD therapy. In this review, we will discuss the application and clinical significance of lipid nanosystem in delivering therapeutic compounds for anti-PD treatment. These medicinal compounds include ropinirole, apomorphine, bromocriptine, astaxanthin, resveratrol, dopamine, glyceryl monooleate, levodopa, N-3,4-bis(pivaloyloxy)- dopamine and fibroblast growth factor, which have significant potential to treat PD in the early stage. This review, in a nutshell, will pave the way for researchers to develop diagnostic and potential therapeutic approaches using nanomedicine to overcome the challenges posed by the BBB in delivering therapeutic compounds for PD.
Collapse
Affiliation(s)
- Subham Preetam
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika, 59053, Sweden; Centre for Biotechnology, Siksha O Anusandhan (SOA-DU), Bhubaneswar 751030, Odisha, India.
| | - Swathi Jonnalagadda
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, India.
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, India.
| | - Rajeswari Rath
- Centre for Biotechnology, Siksha O Anusandhan (SOA-DU), Bhubaneswar 751030, Odisha, India.
| | - Soham Chattopadhyay
- Department of Zoology, Maulana Azad College, Kolkata, Kolkata-700013, West Bengal, India.
| | - Badrah S Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Adel M Abuzenadah
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India.
| | - Akash Gautam
- Centre for Neural and Cognitive Sciences, University of Hyderabad, Hyderabad 500046, India.
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand, 834001, India; Guru Nanak College of Pharmaceutical Sciences, Chakrata Road, Jhajra, Dehradun 248007, India.
| | - Ghulam Md Ashraf
- University of Sharjah, College of Health Sciences, and Research Institute for Medical and Health Sciences, Department of Medical Laboratory Sciences.
| |
Collapse
|
49
|
Rathmacher JA, Fuller JC, Abumrad NN, Flynn CR. Inflammation Biomarker Response to Oral 2-Hydroxybenzylamine (2-HOBA) Acetate in Healthy Humans. Inflammation 2023; 46:1343-1352. [PMID: 36935449 PMCID: PMC10025056 DOI: 10.1007/s10753-023-01801-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/21/2023]
Abstract
Inflammation is associated with the formation of reactive oxygen species (ROS) and the formation of lipid-derived compounds, such as isolevuglandins (IsoLGs), malondialdehyde, 4-hydroxy-nonenal, and 4-oxo-nonenal. The most reactive of these are the IsoLGs, which form covalent adducts with lysine residues and other cellular primary amines leading to changes in protein function, immunogenicity, and epigenetic alterations and have been shown to contribute to a number of inflammatory diseases. 2-Hydroxybenzylamine (2-HOBA) is a natural compound found in buckwheat seeds and reacts with all IsoLG adducts preventing adduct formation with proteins and DNA. Therefore, 2-HOBA is well positioned as an agent for the prevention of inflammatory-prone diseases. In this study, we examined the potential beneficial effects of 2-HOBA on oxidative stress and inflammatory biomarkers in two cohorts of healthy younger and older adults. We utilized the Olink® targeted inflammation panel before and after an oral 15-day treatment regimen with 2-HOBA. We found significant relative changes in the plasma concentration of 15 immune proteins that may reflect the in vivo immune targets of 2-HOBA. Treatment of 2-HOBA resulted in significant increased levels of CCL19, IL-12β, IL-20Rα, and TNFβ, whereas levels of TWEAK significantly decreased. Ingenuity Pathway Analysis identified canonical pathways regulated by the differentially secreted cytokines, chemokines, and growth factors upon 2-HOBA treatment and further points to biofunctions related to the recruitment, attraction, and movement of different immune cell types. In conclusion, 2-HOBA significantly altered the protein biomarkers CCL19, IL-12β, IL-20Rα, TNFβ, and TWEAK, and these may be responsible for the protective effects of 2-HOBA against reactive electrophiles, such as IsoLGs, commonly expressed in conditions of excessive oxidative stress. 2-HOBA has a role as a IsoLG scavenger to proactively improve immune health in a variety of conditions.
Collapse
Affiliation(s)
- John A Rathmacher
- MTI BioTech, Inc, Iowa State University Research Park, Ames, IA, USA
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | | | - Naji N Abumrad
- MTI BioTech, Inc, Iowa State University Research Park, Ames, IA, USA
- Metabolic Technologies, LLC, Missoula, MT, USA
- Department of Surgery, Vanderbilt University Medical Center, MRBIV Room 8465A, Nashville, TN, 37232, USA
| | - Charles R Flynn
- Department of Surgery, Vanderbilt University Medical Center, MRBIV Room 8465A, Nashville, TN, 37232, USA.
| |
Collapse
|
50
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. The central role of the NLRP3 inflammasome pathway in the pathogenesis of age-related diseases in the eye and the brain. Ageing Res Rev 2023; 88:101954. [PMID: 37187367 DOI: 10.1016/j.arr.2023.101954] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
With increasing age, structural changes occur in the eye and brain. Neuronal death, inflammation, vascular disruption, and microglial activation are among many of the pathological changes that can occur during ageing. Furthermore, ageing individuals are at increased risk of developing neurodegenerative diseases in these organs, including Alzheimer's disease (AD), Parkinson's disease (PD), glaucoma and age-related macular degeneration (AMD). Although these diseases pose a significant global public health burden, current treatment options focus on slowing disease progression and symptomatic control rather than targeting underlying causes. Interestingly, recent investigations have proposed an analogous aetiology between age-related diseases in the eye and brain, where a process of chronic low-grade inflammation is implicated. Studies have suggested that patients with AD or PD are also associated with an increased risk of AMD, glaucoma, and cataracts. Moreover, pathognomonic amyloid-β and α-synuclein aggregates, which accumulate in AD and PD, respectively, can be found in ocular parenchyma. In terms of a common molecular pathway that underpins these diseases, the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome is thought to play a vital role in the manifestation of all these diseases. This review summarises the current evidence regarding cellular and molecular changes in the brain and eye with age, similarities between ocular and cerebral age-related diseases, and the role of the NLRP3 inflammasome as a critical mediator of disease propagation in the eye and the brain during ageing.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Moradeke M Adesina
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|