1
|
Li E, Niu W, Lu C, Wang M, Xu X, Xu K, Xu P. Interoception and aging. Ageing Res Rev 2025; 108:102743. [PMID: 40188990 DOI: 10.1016/j.arr.2025.102743] [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: 12/04/2024] [Revised: 03/04/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Interoception refers to the body's perception and regulation of internal physiological states and involves complex neural mechanisms and sensory systems. The current definition of interoception falls short of capturing the breadth of related research; here, we propose an updated definition. Homeostasis, a foundational principle of integrated physiology, is the process by which organisms dynamically maintain optimal balance across all conditions through neural, endocrine, and behavioral functions. This review examines the role of interoception in body homeostasis. Aging is a complex process influenced by multiple factors and involving multiple levels, including physical, psychological, and cognitive. However, interoceptive and aging interoceptive interactions are lacking. A new perspective on interoception and aging holds significant implications for understanding how aging regulates interoception and how interoception affects the aging process. Finally, we summarize that arachidonic acid metabolites show promise as biomarkers of interoception-aging. The aim of this study is to comprehensively analyze interoceptive-aging interactions, understand the aging mechanism from a novel perspective, and provide a theoretical basis for exploring anti-aging strategies.
Collapse
Affiliation(s)
- Erliang Li
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710000, China.
| | - Wenjing Niu
- Changlefang Community Health Service Center, Xi'an 710000, China
| | - Chao Lu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China
| | - Min Wang
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710000, China
| | - Xin Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China
| | - Ke Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China.
| | - Peng Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710000, China.
| |
Collapse
|
2
|
Tang C, Wang J, Ge M, Fu L, Huang J, Yadav H, Shi J, Feng S, Wu F. DSS-induced colitis exacerbates Alzheimer's pathology via neutrophil elastase and cathepsin B activation. Int Immunopharmacol 2025; 155:114666. [PMID: 40228423 DOI: 10.1016/j.intimp.2025.114666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD), the leading cause of dementia, is characterized by amyloid plaques and neuroinflammation, which collectively result in cognitive decline. Peripheral inflammation, particularly intestinal inflammation, has been implicated in exacerbating AD pathology via the gut-brain axis. This study investigated the effects of dextran sulfate sodium (DSS)-induced colitis on amyloid-beta (Aβ) pathology, synaptic integrity, and cognitive function in 5xFAD mice, and explored the roles of neutrophil elastase (NE) and Cathepsin B in these processes. DSS-induced colitis significantly worsened Aβ pathology, evidenced by increased Aβ plaque deposition and elevated soluble Aβ1-42 levels in the brain of 5xFAD mice. The inflammatory state triggered extensive neutrophil infiltration and elevated NE levels in the hippocampus, which were closely associated with Cathepsin B activation. This enzymatic cascade is associated with synaptic damage and cognitive deficits. Treatment with the NE inhibitor Sivelestat effectively suppressed NE-mediated Cathepsin B activation, reduced Aβ pathology, restored dendritic spine density, and improved cognitive performance. Additionally, the Cathepsin B inhibitor CA-074 methyl ester (CA-074Me) mitigated the adverse effects of DSS-induced colitis, further emphasizing the role of Cathepsin B in mediating inflammation-driven AD pathology. These findings reveal that the NE-Cathepsin B axis links peripheral inflammation to exacerbated Aβ pathology, synaptic damage, and cognitive impairment, underscoring the potential of targeting NE and Cathepsin B as therapeutic strategies for inflammation-driven AD progression.
Collapse
Affiliation(s)
- Chong Tang
- Department of General Surgery, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, China; Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226019, Jiangsu, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226019, Jiangsu, China
| | - Minglei Ge
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226019, Jiangsu, China
| | - Li Fu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226019, Jiangsu, China
| | - Jiayue Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226019, Jiangsu, China
| | - Hanshika Yadav
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226019, Jiangsu, China
| | - Jianhua Shi
- Institute for translational neuroscience, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Shichun Feng
- Department of General Surgery, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, China.
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226019, Jiangsu, China.
| |
Collapse
|
3
|
Zhang H, Zhou LQ, Yang S, Dong MH, Chen L, Lu YL, Zhang LY, Zhang L, Chu YH, Xu LL, Pang XW, Zhu LF, Xu T, Yong TY, Wang W, Tian DS, Qin C. The foam cell-derived exosomes exacerbate ischemic white matter injury via transmitting metabolic defects to microglia. Cell Metab 2025:S1550-4131(25)00219-0. [PMID: 40345179 DOI: 10.1016/j.cmet.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/25/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
Atherosclerosis (AS) has been shown to be an independent risk factor for vascular cognitive impairment (VCI), but the mechanisms remain unclear. Here, we found that AS circulating exosomes exacerbated ischemic white matter injury and VCI. Exosomes originating from macrophage-derived foam cells targeted microglia. Mechanistically, foam cell-derived exosomes transmitted redox imbalance, mitochondrial dysfunction, and metabolic defects to microglia via the miR-101-3p-Nrf2-Slc2a1 axis. Anti-miR-101-3p or activation of Nrf2, both genetically and pharmacologically, could antagonize AS exosomes and ameliorate VCI. In conclusion, our findings reveal a distant connection between peripheral macrophages and brain microglia, which provides new insights and potential targets of AS-induced VCI.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi-Lin Lu
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lu-Yang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lan Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lu-Lu Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li-Fang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Xu
- Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Road, Guangzhou 510080, China
| | - Tu-Ying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
4
|
Liu X, Liu Y, Gao Y, Zhang C, Gu C, Lv J, Wu J, Su W. Single-cell profiling unveils a geroprotective role of Procyanidin C1 in hematopoietic immune system via senolytic and senomorphic effects. NPJ AGING 2025; 11:31. [PMID: 40316527 PMCID: PMC12048486 DOI: 10.1038/s41514-025-00222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
Aging of hematopoietic and immune system (HIS) leads to cellular senescence and immune dysregulation, contributing to age-related diseases. Here, we show that Procyanidin C1 (PCC1), a compound with both senolytic and senomorphic properties, can counteract aging-related changes in HIS. Using single-cell RNA sequencing and validation experiments, we found that aging induced cellular senescence, inflammation, and immune dysregulation in the bone marrow and spleen tissues of mice. Long-term PCC1 treatment improved key physiological parameters especially the grip strength of aged mice. Further single-cell analysis revealed PCC1's broad geroprotective effects on HIS, including an increase in the proportion of B cells (BCs) and hematopoietic stem cells (HSCs), suppression of senescence-associated markers, and restoration of normal immune processes. Specifically, PCC1 mitigated inflammation and restored immune homeostasis in BCs by suppressing Cebpb expression and age-associated BCs. Moreover, PCC1 reversed aging-induced alterations in HSCs through upregulating Nedd4 and CD62L-Ca2+ axis expression. Finally, we identified senescent cells (SnCs) using machine learning and gene set enrichment analysis, revealing that PCC1 induced apoptosis of SnCs and regulated their metabolic processes, particularly in granulocytes and myeloid cells. The experimental validation further confirmed the senolytic and senomorphic effects of PCC1 both in vivo and in vitro. Overall, PCC1 holds potential as a therapeutic agent for alleviating immune dysfunction and promoting healthy aging via senolytic and senomorphic effects.
Collapse
Affiliation(s)
- Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yidan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yuehan Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Chun Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chenyang Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jianjie Lv
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Junying Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
5
|
Hu C. Prevention of cardiovascular disease for healthy aging and longevity: A new scoring system and related "mechanisms-hallmarks-biomarkers". Ageing Res Rev 2025; 107:102727. [PMID: 40096912 DOI: 10.1016/j.arr.2025.102727] [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: 05/09/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Healthy "environment-sleep-emotion-exercise-diet" intervention [E(e)SEEDi] lifestyle can improve the quality of life, prolong aging and promote longevity due to improvement of human immunity and prevention of cardiovascular diseases (CVD). Here, the author reviewed the associations between these core elements with CVD and cardiovascular aging, and developed a new scoring system based on the healthy E(e)SEEDi lifestyle for prediction and evaluation of life expectancy. These core factors are assigned 20 points each (120 points in total), and a higher score predicts healthier aging and longevity. The E(e)SEEDi represents "a tree of life" bearing the fruits of longevity as well as "a rocket of anti-ageing" carrying people around the world on a journey of longevity. In conclusion, the E(e)SEEDi can delay aging and increase the life expectancy due to the role of a series of cellular and molecular "mechanisms-hallmarks-biomarkers". It's believed that the novel scoring system has a huge potential and beautiful prospects.
Collapse
Affiliation(s)
- Chunsong Hu
- Department of Cardiovascular Medicine, Nanchang University, Hospital of Nanchang University, Jiangxi Academy of Medical Science, No. 461 Bayi Ave, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
6
|
Heneka MT, van der Flier WM, Jessen F, Hoozemanns J, Thal DR, Boche D, Brosseron F, Teunissen C, Zetterberg H, Jacobs AH, Edison P, Ramirez A, Cruchaga C, Lambert JC, Laza AR, Sanchez-Mut JV, Fischer A, Castro-Gomez S, Stein TD, Kleineidam L, Wagner M, Neher JJ, Cunningham C, Singhrao SK, Prinz M, Glass CK, Schlachetzki JCM, Butovsky O, Kleemann K, De Jaeger PL, Scheiblich H, Brown GC, Landreth G, Moutinho M, Grutzendler J, Gomez-Nicola D, McManus RM, Andreasson K, Ising C, Karabag D, Baker DJ, Liddelow SA, Verkhratsky A, Tansey M, Monsonego A, Aigner L, Dorothée G, Nave KA, Simons M, Constantin G, Rosenzweig N, Pascual A, Petzold GC, Kipnis J, Venegas C, Colonna M, Walter J, Tenner AJ, O'Banion MK, Steinert JR, Feinstein DL, Sastre M, Bhaskar K, Hong S, Schafer DP, Golde T, Ransohoff RM, Morgan D, Breitner J, Mancuso R, Riechers SP. Neuroinflammation in Alzheimer disease. Nat Rev Immunol 2025; 25:321-352. [PMID: 39653749 DOI: 10.1038/s41577-024-01104-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/20/2025]
Abstract
Increasing evidence points to a pivotal role of immune processes in the pathogenesis of Alzheimer disease, which is the most prevalent neurodegenerative and dementia-causing disease of our time. Multiple lines of information provided by experimental, epidemiological, neuropathological and genetic studies suggest a pathological role for innate and adaptive immune activation in this disease. Here, we review the cell types and pathological mechanisms involved in disease development as well as the influence of genetics and lifestyle factors. Given the decade-long preclinical stage of Alzheimer disease, these mechanisms and their interactions are driving forces behind the spread and progression of the disease. The identification of treatment opportunities will require a precise understanding of the cells and mechanisms involved as well as a clear definition of their temporal and topographical nature. We will also discuss new therapeutic strategies for targeting neuroinflammation, which are now entering the clinic and showing promise for patients.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg.
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Jeroen Hoozemanns
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Dietmar Rudolf Thal
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Charlotte Teunissen
- Department of Laboratory Medicine, VUMC Amsterdam, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Imperial College London, London, UK
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Washington School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Agustin Ruiz Laza
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Jose Vicente Sanchez-Mut
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Alicante, Spain
| | - Andre Fischer
- Clinic for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
- Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Disease (DZNE), Göttingen, Germany
| | - Sergio Castro-Gomez
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Michael Wagner
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center Munich, Biochemistry, Medical Faculty, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Sim K Singhrao
- Brain and Behaviour Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian Kleemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip L De Jaeger
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Hannah Scheiblich
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Guy C Brown
- Deparment of Biochemistry, University of Cambridge, Cambridge, UK
| | - Gary Landreth
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Miguel Moutinho
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Ising
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Karabag
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Darren J Baker
- Department of Paediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York City, NY, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Malu Tansey
- College of Medicine, University of Florida, Gainsville, FL, USA
| | - Alon Monsonego
- Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Guillaume Dorothée
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Hôpital Saint-Antoine, Paris, France
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Neta Rosenzweig
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University of Bonn, Bonn, Germany
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto Biosanitario de Granada (ibs.Granada), Granada, Spain
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jochen Walter
- Center of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behaviour, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Douglas L Feinstein
- Department of NeuroAnesthesia, University of Illinois at Chicago, Chicago, IL, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kiran Bhaskar
- Department of Molecular Genetics & Microbiology and Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Todd Golde
- Department of Pharmacology and Chemical Biology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | | | - David Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John Breitner
- Department of Psychiatry, McGill University Faculty of Medicine, Montreal, Québec, Canada
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sean-Patrick Riechers
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
| |
Collapse
|
7
|
Zhang Z, Yang R, Zi Z, Liu B. A new clinical age of aging research. Trends Endocrinol Metab 2025; 36:440-458. [PMID: 39227191 DOI: 10.1016/j.tem.2024.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024]
Abstract
Aging is a major risk factor for a variety of diseases, thus, translation of aging research into practical applications is driven by the unmet need for existing clinical therapeutic options. Basic and translational research efforts are converging at a critical stage, yielding insights into how fundamental aging mechanisms are used to identify promising geroprotectors or therapeutics. This review highlights several research areas from a clinical perspective, including senescent cell targeting, alleviation of inflammaging, and optimization of metabolism with endogenous metabolites or precursors. Refining our understanding of these key areas, especially from the clinical angle, may help us to better understand and attenuate aging processes and improve overall health outcomes.
Collapse
Affiliation(s)
- Zhen Zhang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518055, China
| | - Renlei Yang
- Department of Plastic Surgery, Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Zhike Zi
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518055, China.
| |
Collapse
|
8
|
Hansen CE, Konings J, Toth G, Chornyi S, Karsten M, van Het Hof B, van der Pol SMA, Beekhuis-Hoekstra SD, Kok N, Fung WK, Dijksman NS, Baron W, Witte ME, Lanekoff I, de Vries HE, Kooij G. Spatial mapping of the AA-PGE 2-EP axis in multiple sclerosis lesions. Acta Neuropathol 2025; 149:39. [PMID: 40299057 PMCID: PMC12041062 DOI: 10.1007/s00401-025-02878-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
Bioactive lipid mediators (LMs) derived from polyunsaturated fatty acids (PUFAs) are key molecules in both the initiation and resolution of inflammatory responses. Previous findings suggest that a dysregulated LM balance, especially within the arachidonic acid (AA) pathway, may contribute to an impaired resolution response and subsequent chronic neuroinflammation in multiple sclerosis (MS). However, to date, the local biosynthesis and signaling of LMs within the brain of people with MS (PwMS) remains unexplored. In this study, we, therefore, mapped the distribution of AA and its key downstream LM prostaglandin E2 (PGE2) in white matter MS brain tissue and of non-neurological controls (NNCs) for the first time using mass spectrometry imaging. We found that AA levels are lower in MS cases compared to NNCs and reduced in MS lesions compared to peri-lesional tissue. Furthermore, the PGE2/AA ratio, indicating the PGE2 synthesis from the AA substrate, was increased in lesion areas compared to fully myelinated regions in MS. In line with that, the expression of prostaglandin synthesizing enzymes as measured by RT-qPCR was partially increased in MS tissue compared to NNCs. In addition, the expression of prostaglandin E2 receptor 4 (EP4) decreased, while prostaglandin E2 receptor 2 (EP2) showed increased expression levels in MS lesions compared to NNCs and localized specifically to microglia. We also found that PGE2 addition to pro-inflammatory human-induced pluripotent stem cell (iPSC)-derived microglia resulted in enhanced cytokine signaling pathways, but also the upregulation of its synthase PTGES and homeostatic/resolving signaling, the latter of which might mainly occur through EP2 signaling. Collectively, our results provide detailed information about the region-specific levels of AA and PGE2 in MS lesions and we propose enhanced PGE2-EP2 signaling in inflamed microglia in MS.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Julia Konings
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Gabor Toth
- Department of Chemistry, BMC, Uppsala University, 75237, Uppsala, Sweden
- Center of Excellence for the Chemical Mechanisms of Life, Uppsala University, Uppsala, Sweden
| | - Serhii Chornyi
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Manon Karsten
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Susanne M A van der Pol
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Stephanie D Beekhuis-Hoekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Nine Kok
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Wing Ka Fung
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Naomi S Dijksman
- Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, UMCG, MS Center Noord Nederland, A. Deusinglaan 1, Groningen, The Netherlands
| | - Wia Baron
- Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, UMCG, MS Center Noord Nederland, A. Deusinglaan 1, Groningen, The Netherlands
| | - Maarten E Witte
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ingela Lanekoff
- Department of Chemistry, BMC, Uppsala University, 75237, Uppsala, Sweden
- Center of Excellence for the Chemical Mechanisms of Life, Uppsala University, Uppsala, Sweden
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, UMC Location VU Medical Center, Amsterdam, The Netherlands.
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Kang A, Eor JY, Lee J, Kwak MJ, Lee DJ, Seo E, Lee WJ, Son SH, Song M, Kim JM, Kim HW, Yang J, Oh S, Kim Y. Lacticaseibacillus casei IDCC 3451 alleviates cognitive and behavioral functions by reshaping the gut microbiome and regulating intestinal barrier integrity in chronic stress animal models. Curr Res Food Sci 2025; 10:101051. [PMID: 40290371 PMCID: PMC12023876 DOI: 10.1016/j.crfs.2025.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Lacticaseibacillus casei IDCC 3451 (3451) was evaluated for its effects on the gut-brain axis using Caenorhabditis elegans (C. elegans) and mouse models of stress and inflammation. In C. elegans, 3451 extended lifespans by 25 %, improved motility, and chemotaxis, enhanced survival under pathogen challenge, and reduced amyloid beta accumulation by 42 %. Transcriptomic profiling revealed upregulation of genes involved in neurotransmitter signaling and serine/threonine pathways. In the unpredictable chronic mild stress (UCMS) mouse model, 3451 administration increased the time spent in the center of the open field by 65 % and reduced immobility in the forced swim test by 32 %, indicating anxiolytic and antidepressant effects. Serum levels of aspartate aminotransferase (AST) and gamma-glutamyl transferase (GGT) were decreased by 18 % and 24 %, respectively. Additionally, 3451 restored the expressions of 5HT1AR, GABAR, and tight junction proteins, including ZO-1 and Claudin1. Metabolomic analysis showed increased glycine and decreased palmitic acid levels, associated with an increased abundance of Ruminococcus and Akkermansia. In the dextran sulfate sodium (DSS)-induced colitis model, 3451 reduced the disease activity index by 36 %, improved colon histology, increased goblet cell preservation, and upregulated ZO-1 and IL-10 expression. Threonine levels were also increased and correlated with a higher abundance of Coprococcus. These findings demonstrate that 3451 improved behavioral and intestinal outcomes through coordinated modulation of host signaling, metabolite production, and gut microbial composition, highlighting its therapeutic potential for managing IBD and neurobehavioral disorders.
Collapse
Affiliation(s)
- Anna Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Ju Young Eor
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Junbeom Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Daniel Junpyo Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Eunsol Seo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Woong Ji Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Seon-hui Son
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, 34134, South Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Gyeonggi-do, South Korea
| | - Hyung Wook Kim
- College of Life Sciences, Sejong University, Seoul, 05006, South Korea
| | - Jungwoo Yang
- Department of Microbiology, College of Medicine, Dongguk University, Gyeongju, 38066, South Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, 55069, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| |
Collapse
|
10
|
Chen W, Wang X, Huang G, Sheng Q, Zhou E. Identification of cellular senescence-related genes as biomarkers for lupus nephritis based on bioinformatics. Front Genet 2025; 16:1551450. [PMID: 40290492 PMCID: PMC12021929 DOI: 10.3389/fgene.2025.1551450] [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] [Received: 12/25/2024] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background Lupus nephritis (LN) is one of the most common and severe complications of systemic lupus erythematosus with unclear pathogenesis. The most accurate diagnosis criterion of LN is still renal biopsy and nowadays treatment strategies of LN are far from satisfactory. Cellular senescence is defined as the permanent cell cycle arrest marked by senescence-associated secretory phenotype (SASP), which has been proved to accelerate the mobility and mortality of patients with LN. The study is aimed to identify cellular senescence-related genes for LN. Methods Genes related to cellular senescence and LN were obtained from the MSigDB genetic database and GEO database respectively. Through differential gene analysis, Weighted Gene Go-expression Network Analysis (WGCNA) and machine learning algorithms, hub cellular senescence-related differentially expressed genes (CS-DEGs) were identified. By external validation, hub CS-DEGs were further filtered and the remaining genes were identified as biomarkers. We explored their potential physiopathologic function through GSEA. Results We obtained 432 genes related to cellular senescence, 1,208 differentially expressed genes (DEGs) and 840 genes in the key gene module related to LN, which were intersected with each other for CS-DEGs. Subsequent Machine learning algorithms screened out six hub CS-DEGs and finally three hub CS-DEGs, ALOX5, PTGER2 and PRKCB passed through external validation, which were identified as biomarkers. The three biomarkers were enriched in "B Cell receptor signaling pathway" and "NF-kappa B signaling pathway" based on GESA results. Conclusion This study explored the potential relationship between cellular senescence and LN, and identified three biomarkers ALOX5, PTGER2, and PRKCB playing key roles in LN, which will provide new insights for the diagnosis and treatment of LN.
Collapse
Affiliation(s)
- Wei Chen
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, Jiangsu, China
- Jiangsu University Key Laboratory of Tonifying Kidney and Anti-senescence, Nanjing, Jiangsu, China
| | - Xiaofang Wang
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, Jiangsu, China
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, China
| | - Guoshun Huang
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, Jiangsu, China
- Jiangsu University Key Laboratory of Tonifying Kidney and Anti-senescence, Nanjing, Jiangsu, China
| | - Qin Sheng
- Department of Nephrology, Suzhou Affiliated Hospital of Nanjing University of Chinese Medicine (Suzhou Hospital of Traditional Chinese Medicine), Suzhou, Jiangsu, China
| | - Enchao Zhou
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, Jiangsu, China
- Jiangsu University Key Laboratory of Tonifying Kidney and Anti-senescence, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Wang J, Gong P, Liu Q, Wang M, Wu D, Li M, Zheng S, Wang H, Long Q. Stimulation of regulatory dendritic cells suppresses cytotoxic T cell function and alleviates DEN-induced liver injury, fibrosis and hepatocellular carcinoma. Front Immunol 2025; 16:1565486. [PMID: 40264769 PMCID: PMC12011597 DOI: 10.3389/fimmu.2025.1565486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
Background Dendritic cells (DCs) are versatile professional antigen-presenting cells and play an instrumental role in the generation of antigen-specific T-cell responses. Modulation of DC function holds promise as an effective strategy to improve anti-tumor immunotherapy efficacy and enhance self-antigen tolerance in autoimmune diseases. Methods Wild-type (WT) and TLR2 knockout (KO) mice at 2 weeks of age were injected intraperitoneally (i.p.) with a single dose of diethylnitrosamine (DEN) to induce hepatocellular carcinoma (HCC). Four weeks later, WT and KO mice were randomly divided into control and treatment groups and treated once every two days for 30 weeks with phosphate buffered saline (PBS) and a mix of 4 TLR2-activating lactic acid-producing probiotics (LAP), respectively. Mice were euthanized after 30 weeks of LAP treatment and their liver tissues were collected for gene expression, histological, flow cytometric and single-cell RNA sequencing analyses. Results We demonstrate here that oral administration of a mix of TLR2-activating LAP triggers a marked accumulation of regulatory DCs (rDCs) in the liver of mice. LAP-treated mice are protected from DEN-induced liver injury, fibrosis and HCC in a TLR2-dependent manner. Single-cell transcriptome profiling revealed that LAP treatment determines an immunosuppressive hepatic T-cell program that is characterized by a significantly reduced cytotoxic activity. The observed functional changes of T cells correlated well with the presence of a hepatic DC subset displaying a regulatory or tolerogenic transcriptional signature. Conclusion Overall, these data suggest that stimulation of regulatory dendritic cells (rDCs) in the liver by LAP suppresses cytotoxic T-cell function and alleviates DEN-induced liver damage, fibrosis and tumorigenesis.
Collapse
Affiliation(s)
- Junjie Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Pixu Gong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qingqing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Menglei Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Dengfang Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Mengyu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Shujie Zheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qiaoming Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
12
|
Boucher DM, Robichaud S, Lorant V, Leon JS, Suliman I, Rasheed A, Susser LI, Emerton C, Geoffrion M, De Jong E, Bowdish DM, Aikawa M, Aikawa E, Singh SA, Rayner KJ, Ouimet M. Age-Related Impairments in Immune Cell Efferocytosis and Autophagy Hinder Atherosclerosis Regression. Arterioscler Thromb Vasc Biol 2025; 45:481-495. [PMID: 39945065 PMCID: PMC11936474 DOI: 10.1161/atvbaha.124.321662] [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: 08/12/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Aging is a well-established risk factor for the development and progression of atherosclerosis, but the molecular mechanisms underlying this relationship remain poorly defined, and its role in atherosclerosis regression is unknown. To uncover age-related alterations that may impair atherosclerosis regression, we investigated the response of young and old macrophages to atherogenic lipoproteins in vitro and in vivo. METHODS Metabolic and proteomic studies were performed in vitro using macrophages differentiated from the bone marrow of young or old mice. To test the role of immune cell aging in atherosclerosis regression, bone marrow from young and old donors was transplanted into irradiated young recipient mice expressing gain-of-function AAV-PCSK9 (adeno-associated virus-proprotein convertase subtilisin/kexin type 9). Following 14 weeks of Western diet feeding, atherosclerosis regression was induced by switching to a standard laboratory diet for 4 weeks. RESULTS Compared with young macrophages, old macrophages accumulated more lipid droplets upon lipid loading with the pro-atherogenic lipoprotein aggregated LDL (low-density lipoprotein), accompanied by a failure to proportionally induce autophagy and cholesterol efflux. Proteomic analysis of bone marrow-derived macrophages revealed that pathways related to endocytosis, engulfment, and phagocytosis were downregulated in old lipid-loaded macrophages. Functional studies confirmed a reduction in efferocytic capacity in old macrophages. In recipient mice transplanted with old bone marrow, atherosclerosis regression was impaired, as evidenced by inefficient resolution of circulating inflammatory cell levels, reduced activation of plaque autophagy and apoptotic cell clearance, and persistent plaque CD45+ and CD68+ content. CONCLUSIONS Aging impairs macrophage function through reduced efferocytosis and autophagy activation, limiting atherosclerosis regression. These results highlight the need to better define the mechanisms linking aging to atherosclerosis to develop targeted therapies for the aging population.
Collapse
Affiliation(s)
- Dominique M. Boucher
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Sabrina Robichaud
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Victoria Lorant
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Jonathan S. Leon
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Issraa Suliman
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Adil Rasheed
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Leah I. Susser
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Christina Emerton
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Erica De Jong
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada (E.D.J., D.M.E.B.)
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, ON, Canada (E.D.J., D.M.E.B.)
| | - Dawn M.E. Bowdish
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada (E.D.J., D.M.E.B.)
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, ON, Canada (E.D.J., D.M.E.B.)
| | - Masanori Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sasha A. Singh
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Katey J. Rayner
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| |
Collapse
|
13
|
Vujosevic S, Limoli C, Kozak I. Hallmarks of aging in age-related macular degeneration and age-related neurological disorders: novel insights into common mechanisms and clinical relevance. Eye (Lond) 2025; 39:845-859. [PMID: 39289517 PMCID: PMC11933422 DOI: 10.1038/s41433-024-03341-5] [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: 04/14/2024] [Revised: 08/13/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Age-related macular degeneration (AMD) and age-related neurological diseases (ANDs), such as Alzheimer's and Parkinson's Diseases, are increasingly prevalent conditions that significantly contribute to global morbidity, disability, and mortality. The retina, as an accessible part of the central nervous system (CNS), provides a unique window to study brain aging and neurodegeneration. By examining the associations between AMD and ANDs, this review aims to highlight novel insights into fundamental mechanisms of aging and their role in neurodegenerative disease progression. This review integrates knowledge from the emerging field of aging research, which identifies common denominators of biological aging, specifically loss of proteostasis, impaired macroautophagy, mitochondrial dysfunction, and inflammation. Finally, we emphasize the clinical relevance of these pathways and the potential for cross-disease therapies that target common aging hallmarks. Identifying these shared pathways could open avenues to develop therapeutic strategies targeting mechanisms common to multiple degenerative diseases, potentially attenuating disease progression and promoting the healthspan.
Collapse
Affiliation(s)
- Stela Vujosevic
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
- Eye Clinic, IRCCS MultiMedica, Milan, Italy.
| | - Celeste Limoli
- Eye Clinic, IRCCS MultiMedica, Milan, Italy
- University of Milan, Milan, Italy
| | - Igor Kozak
- Moorfields Eye Hospital Centre, Abu Dhabi, UAE
- Ophthalmology and Vision Science, University of Arizona, Tucson, USA
| |
Collapse
|
14
|
Jung ES, Choi H, Mook-Jung I. Decoding microglial immunometabolism: a new frontier in Alzheimer's disease research. Mol Neurodegener 2025; 20:37. [PMID: 40149001 PMCID: PMC11948825 DOI: 10.1186/s13024-025-00825-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) involves a dynamic interaction between neuroinflammation and metabolic dysregulation, where microglia play a central role. These immune cells undergo metabolic reprogramming in response to AD-related pathology, with key genes such as TREM2, APOE, and HIF-1α orchestrating these processes. Microglial metabolism adapts to environmental stimuli, shifting between oxidative phosphorylation and glycolysis. Hexokinase-2 facilitates glycolytic flux, while AMPK acts as an energy sensor, coordinating lipid and glucose metabolism. TREM2 and APOE regulate microglial lipid homeostasis, influencing Aβ clearance and immune responses. LPL and ABCA7, both associated with AD risk, modulate lipid processing and cholesterol transport, linking lipid metabolism to neurodegeneration. PPARG further supports lipid metabolism by regulating microglial inflammatory responses. Amino acid metabolism also contributes to microglial function. Indoleamine 2,3-dioxygenase controls the kynurenine pathway, producing neurotoxic metabolites linked to AD pathology. Additionally, glucose-6-phosphate dehydrogenase regulates the pentose phosphate pathway, maintaining redox balance and immune activation. Dysregulated glucose and lipid metabolism, influenced by genetic variants such as APOE4, impair microglial responses and exacerbate AD progression. Recent findings highlight the interplay between metabolic regulators like REV-ERBα, which modulates lipid metabolism and inflammation, and Syk, which influences immune responses and Aβ clearance. These insights offer promising therapeutic targets, including strategies aimed at HIF-1α modulation, which could restore microglial function depending on disease stage. By integrating metabolic, immune, and genetic factors, this review underscores the importance of microglial immunometabolism in AD. Targeting key metabolic pathways could provide novel therapeutic strategies for mitigating neuroinflammation and restoring microglial function, ultimately paving the way for innovative treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- Eun Sun Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Hayoung Choi
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Inhee Mook-Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Korea Dementia Research Center, Seoul, South Korea.
| |
Collapse
|
15
|
Mosharov EV, Rosenberg AM, Monzel AS, Osto CA, Stiles L, Rosoklija GB, Dwork AJ, Bindra S, Junker A, Zhang Y, Fujita M, Mariani MB, Bakalian M, Sulzer D, De Jager PL, Menon V, Shirihai OS, Mann JJ, Underwood MD, Boldrini M, Thiebaut de Schotten M, Picard M. A human brain map of mitochondrial respiratory capacity and diversity. Nature 2025:10.1038/s41586-025-08740-6. [PMID: 40140564 DOI: 10.1038/s41586-025-08740-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 02/04/2025] [Indexed: 03/28/2025]
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) powers brain activity1,2, and mitochondrial defects are linked to neurodegenerative and neuropsychiatric disorders3,4. To understand the basis of brain activity and behaviour, there is a need to define the molecular energetic landscape of the brain5-10. Here, to bridge the scale gap between cognitive neuroscience and cell biology, we developed a physical voxelization approach to partition a frozen human coronal hemisphere section into 703 voxels comparable to neuroimaging resolution (3 × 3 × 3 mm). In each cortical and subcortical brain voxel, we profiled mitochondrial phenotypes, including OXPHOS enzyme activities, mitochondrial DNA and volume density, and mitochondria-specific respiratory capacity. We show that the human brain contains diverse mitochondrial phenotypes driven by both topology and cell types. Compared with white matter, grey matter contains >50% more mitochondria. Moreover, the mitochondria in grey matter are biochemically optimized for energy transformation, particularly among recently evolved cortical brain regions. Scaling these data to the whole brain, we created a backwards linear regression model that integrates several neuroimaging modalities11 to generate a brain-wide map of mitochondrial distribution and specialization. This model predicted mitochondrial characteristics in an independent brain region of the same donor brain. This approach and the resulting MitoBrainMap of mitochondrial phenotypes provide a foundation for exploring the molecular energetic landscape that enables normal brain function. This resource also relates to neuroimaging data and defines the subcellular basis for regionalized brain processes relevant to neuropsychiatric and neurodegenerative disorders. All data are available at http://humanmitobrainmap.bcblab.com .
Collapse
Affiliation(s)
- Eugene V Mosharov
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Ayelet M Rosenberg
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S Monzel
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Corey A Osto
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gorazd B Rosoklija
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew J Dwork
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Snehal Bindra
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Alex Junker
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Madeline B Mariani
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mihran Bakalian
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Departments of Neurology and Pharmacology, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - J John Mann
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mark D Underwood
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Maura Boldrini
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behaviour Laboratory, Paris, France.
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA University of Bordeaux, Bordeaux, France.
| | - Martin Picard
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, USA.
- Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
16
|
Prasongtanakij S, Soontrapa K, Thumkeo D. The role of prostanoids in regulatory T cells and their implications in inflammatory diseases and cancers. Eur J Cell Biol 2025; 104:151482. [PMID: 40184828 DOI: 10.1016/j.ejcb.2025.151482] [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: 12/06/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025] Open
Abstract
Regulatory T cells (Tregs) play an important role in the immune system through the regulation of immunological self-tolerance and homeostasis. Furthermore, increasing evidence suggests the potential contribution of Tregs beyond immunity in the process of repairing various injured tissues. Tregs are generally characterised by the constitutive expression of forkhead box protein 3 (FOXP3) transcription factor in the nucleus and high expression levels of CD25 and CTLA-4 on the cell surface. To date, a large number of molecules have been identified as key regulators of Treg differentiation and function. Among these molecules are prostanoids, which are multifaceted lipid mediators. Prostanoids are produced from arachidonic acid through the catalytic activity of the enzyme cyclooxygenase and exert their functions through the 9 cognate receptors, DP1-2, EP1-EP4, FP, IP and TP. We briefly review previous studies on the regulatory mechanism of Tregs and then discuss recent works on the modulatory role of prostanoids.
Collapse
Affiliation(s)
- Somsak Prasongtanakij
- Laboratory of Immunopharmacology, Kyoto University Graduate School of Medicine, Japan
| | - Kitipong Soontrapa
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Dean Thumkeo
- Laboratory of Immunopharmacology, Kyoto University Graduate School of Medicine, Japan; Center for Medical Education and Internationalization, Kyoto University Faculty of Medicine, Japan.
| |
Collapse
|
17
|
Xie Z, Lin M, Xing B, Wang H, Zhang H, Cai Z, Mei X, Zhu ZJ. Citrulline regulates macrophage metabolism and inflammation to counter aging in mice. SCIENCE ADVANCES 2025; 11:eads4957. [PMID: 40053596 PMCID: PMC11887811 DOI: 10.1126/sciadv.ads4957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Metabolic dysregulation and altered metabolite concentrations are widely recognized as key characteristics of aging. Comprehensive exploration of endogenous metabolites that drive aging remains insufficient. Here, we conducted an untargeted metabolomics analysis of aging mice, revealing citrulline as a consistently down-regulated metabolite associated with aging. Systematic investigations demonstrated that citrulline exhibited antiaging effects by reducing cellular senescence, protecting against DNA damage, preventing cell cycle arrest, modulating macrophage metabolism, and mitigating inflammaging. Long-term citrulline supplementation in aged mice yielded beneficial effects and ameliorated age-associated phenotypes. We further elucidated that citrulline acts as an endogenous metabolite antagonist to inflammation, suppressing proinflammatory responses in macrophages. Mechanistically, citrulline served as a potential inhibitor of mammalian target of rapamycin (mTOR) activation in macrophage and regulated the mTOR-hypoxia-inducible factor 1α-glycolysis signaling pathway to counter inflammation and aging. These findings underscore the significance of citrulline deficiency as a driver of aging, highlighting citrulline supplementation as a promising therapeutic intervention to counteract aging-related changes.
Collapse
Affiliation(s)
- Zhangdan Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Moubin Lin
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Beizi Xing
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongmiao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Haosong Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zimu Cai
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, P. R. China
| | - Xinyu Mei
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, P.R. China
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Shanghai Key Laboratory of Aging Studies, Shanghai 201210, P. R. China
| |
Collapse
|
18
|
Newman PP, Schmitt BL, Maurmann RM, Pence BD. Polysaccharides with Arabinose: Key Players in Reducing Chronic Inflammation and Enhancing Immune Health in Aging. Molecules 2025; 30:1178. [PMID: 40076400 PMCID: PMC11901799 DOI: 10.3390/molecules30051178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Aging is associated with a decline in physiological performance leading to increased inflammation and impaired immune function. Polysaccharides (PLs) found in plants, fruits, and fungi are emerging as potential targets for therapeutic intervention, but little is known about their effects on chronic inflammation and aging. This review aims to highlight the current advances related to the use of PLs, with the presence of arabinose, to attenuate oxidative stress and chronic and acute inflammation, and their immunomodulatory effects associated with antioxidant status in monocytes, macrophages, and neutrophil infiltration, and leukocyte rolling adhesion in neutrophils. In addition, recent studies have shown the importance of investigating the 'major' monosaccharide, such as arabinose, present in several of these polysaccharides, and with described effects on gut microbiome, glucose, inflammation, allergy, cancer cell proliferation, neuromodulation, and metabolic stress. Perspectives and opportunities for further investigation are provided. By promoting a balanced immune response and reducing inflammation, PLs with arabinose or even arabinose per se may alleviate the immune dysregulation and inflammation seen in the elderly, therefore providing a promising strategy to mitigate a variety of diseases.
Collapse
Affiliation(s)
- Patricia Pantoja Newman
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA; (B.L.S.); (R.M.M.)
| | | | | | - Brandt D. Pence
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA; (B.L.S.); (R.M.M.)
| |
Collapse
|
19
|
Fiala M, Hammock BD, Hwang SH, Whitelegge J, Paul K, Kaczor-Urbanowicz KE, Urbanowicz A, Kesari S. Inhibitors of soluble epoxide hydrolase and cGAS/STING repair defects in amyloid-β clearance underlying vascular complications of Alzheimer's disease. J Alzheimers Dis 2025; 104:150-157. [PMID: 39962970 PMCID: PMC11969680 DOI: 10.1177/13872877241305965] [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] [Indexed: 03/15/2025]
Abstract
BackgroundAlzheimer's disease (AD) and its monoclonal antibody therapies are associated with brain vasculitis and amyloid-related imaging abnormalities. The naturally-formed epoxides (EpFAs) of polyunsaturated fatty acids (PUFAs), such as 11,12-epoxyeicosatetraenoic acid (EEQ), are anti-inflammatory and pro-resolution mediators, which are increased by dietary supplementation with ω-3 PUFAs. EpFAs are, however, enzymatically hydrolyzed by soluble epoxide hydrolase (sEH) in AD patients' macrophages in vivo and in vitro.ObjectiveTo repair amyloid-β 1-42 (Aβ) degradation by AD macrophages using the inhibitors of a) soluble epoxide hydrolase (sEHIs), termed TPPU and EC5026, together with EpFAs, or b) STING pathway termed H-151.MethodsImmunobiology, immunochemistry, RNA sequencing, and confocal microscopy were used.ResultsIn AD brain (examined postmortem), monocyte/macrophages upload Aβ in plaques and transfer it without degradation into brain microvessels, suffer apoptotis, and release Aβ, inducing vasculitis. The EpFAs of epoxyeicosatetraenoic acid (EEQ), along with the inhibitors TPPU and H-151, decrease inflammatory cytokines and regulate macrophage unfolded protein response to endoplasmic reticulum stress. Treatment of AD macrophages by TPPU with EEQ or by STING inhibitor H-151 increased uploading of Aβ after 2 hours and increased degradation of Aβ after 24 hours.ConclusionsThe sEHI inhibitor EC5026 and the STING inhibitor H-151 increased macrophage uptake and degradation of Aβ. EC5026 administration was safe in normal volunteers. EC5026 together with ω-3 PUFA supplementation are indicated for in a clinical trial in patients with mild cognitive impairment.
Collapse
Affiliation(s)
- Milan Fiala
- Department of Integrative Biology and Physiology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center University of California-Davis, Davis, CA, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center University of California-Davis, Davis, CA, USA
| | - Julian Whitelegge
- Department of Psychiatry, UCLA School of Medicine, Los Angeles, CA, USA
| | - Ketema Paul
- Department of Integrative Biology and Physiology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Center for Oral and Head/Neck Oncology Research, Biosystems and Function, UCLA School of Dentistry, University of California at Los Angeles, Los Angeles, CA, USA
- UCLA Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, CA, USA
- Institute of Control and Computation Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Andrzej Urbanowicz
- Center for Oral and Head/Neck Oncology Research, Biosystems and Function, UCLA School of Dentistry, University of California at Los Angeles, Los Angeles, CA, USA
- UCLA Institute for Quantitative and Computational Biosciences, University of California at Los Angeles, Los Angeles, CA, USA
- Institute of Control and Computation Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Santosh Kesari
- Providence Saint John’s Health Center, Santa Monica, CA, USA
| |
Collapse
|
20
|
Lucassen PJ, Korosi A, de Rooij SR, Smit AB, Van Dam AM, Daskalakis NP, Van Kesteren RE, Verheijen MHG, Lesuis SL, Kessels HW, Krugers HJ. How Can Early Stress Influence Later Alzheimer's Disease Risk? Possible Mediators and Underlying Mechanisms. Biol Psychiatry 2025; 97:372-381. [PMID: 39577793 DOI: 10.1016/j.biopsych.2024.11.007] [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: 07/25/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Alzheimer's disease (AD) is a progressive, age-related neurodegenerative disorder to which genetic mutations and risk factors contribute. Evidence is increasing that environmental and lifestyle-related factors, such as exercise, nutrition, education, and exposure to (early-life) stress modify the onset, incidence, and progression of AD. Here, we discuss recent preclinical findings on putative substrates that can explain or contribute to the effects of stress early in life on the risk of developing AD. We focus in particular on stress hormones, neural networks, synapses, mitochondria, nutrient and lipid metabolism, adult neurogenesis, engram cell ensembles, and neuroinflammation. We discuss the idea that stress exposure early in life can alter these processes, either combined or in isolation, thereby reducing the capacity of the brain to resist deleterious consequences of, for example, amyloid-β accumulation, thereby accelerating cognitive decline and progression of Alzheimer-related changes in model systems of the disease. A better understanding of whether experiences early in life also modify trajectories of cognitive decline and pathology in AD and how the substrates discussed translate to humans may help develop novel preventive and/or therapeutic strategies to mitigate the consequences of stressors early in life and increase resilience to developing dementia.
Collapse
Affiliation(s)
- Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands.
| | - Aniko Korosi
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Anne-Marie Van Dam
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Nikolaos P Daskalakis
- Neurogenomics and Translational Bioinformatics Laboratory, McLean Hospital, Harvard University, Boston, Massachusetts
| | - Ronald E Van Kesteren
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sylvie L Lesuis
- Department of Cellular & Computational Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Helmut W Kessels
- Department of Cellular & Computational Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
21
|
Rodríguez JP, Casas J, Balboa MA, Balsinde J. Bioactive lipid signaling and lipidomics in macrophage polarization: Impact on inflammation and immune regulation. Front Immunol 2025; 16:1550500. [PMID: 40028333 PMCID: PMC11867965 DOI: 10.3389/fimmu.2025.1550500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Macrophages, crucial innate immune cells, defend against pathogens and resolve inflammation, maintaining tissue balance. They perform phagocytosis, present antigens to T cells, and bond innate and adaptive immunity through various activation states. Classical activation is associated with Th1 responses and interferon γ production, while alternative activation, induced by interleukin 4, is characterized by increased endocytosis, reduced secretion of pro-inflammatory cytokines, and roles in immunoregulation and tissue remodeling. Although these represent opposite extremes observed in vitro, the remarkable plasticity of macrophages allows for a wide spectrum of activation phenotypes that are complex to characterize experimentally. While the application of omics techniques has resulted in significant advances in the characterization of macrophage polarization, lipidomic studies have received lesser attention. Beyond their role as structural components and energy sources, lipids function as signaling molecules that regulate macrophage activation and polarization, thereby shaping immune responses. This work reviews the interaction between lipid signaling and macrophage polarization, exploring how lipid metabolism influences macrophage phenotype and function. These insights offer potential therapeutic strategies for immune-mediated diseases and inflammation-related disorders, including inflammaging.
Collapse
Affiliation(s)
- Juan P. Rodríguez
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del Nordeste Argentino (IQUIBA-NEA), Universidad Nacional del Nordeste, Consejo Nacional de Investigaciones Científicas y Técnicas (UNNE-CONICET), Corrientes, Argentina
| | - Javier Casas
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Valladolid, Spain
| | - María A. Balboa
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Balsinde
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Beltran-Velasco AI. Brain Glycogen-Its Metabolic Role in Neuronal Health and Neurological Disorders-An Extensive Narrative Review. Metabolites 2025; 15:128. [PMID: 39997753 PMCID: PMC11857135 DOI: 10.3390/metabo15020128] [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: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Brain glycogen is imperative for neuronal health, as it supports energy demands and metabolic processes. This review examines the pathways involved in glycogen storage and utilization in the central nervous system, emphasizing their role in both physiology and pathology. It explores how alterations in glycogen metabolism contribute to neurological disorders, including neurodegenerative diseases, epilepsy, and metabolic conditions while highlighting the bidirectional interaction between neurons and glia in maintaining brain homeostasis. Methods: A comprehensive search of articles published between 2015 and 2025 was conducted using the following databases: ScienceDirect, Scopus, Wiley, Web of Science, Medline, and PubMed. The selection of relevant studies was based on their focus on brain glycogen metabolism and its role in neurological conditions, with studies that did not meet the inclusion criteria being excluded. Results: The metabolic processes of brain glycogen are subject to rigorous regulation by astrocyte-neuron interactions, thereby ensuring metabolic homeostasis and energy availability. The dysregulation of glycogen storage and mobilization has been implicated in the development of synaptic dysfunction, excitotoxicity, and neurodegeneration in a variety of disorders. For instance, aberrant glycogen accumulation in diseases such as Lafora disease has been associated with severe neurodegeneration, while impaired glycogen mobilization has been shown to exacerbate energy deficits in Alzheimer's and epilepsy. Conclusions: Targeting brain glycogen metabolism represents a promising approach for therapeutic intervention in neurological disorders. However, the translation of these strategies to human models remains challenging, particularly with regard to the long-term safety and specificity of glycogen-targeted therapies.
Collapse
|
23
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025; 16:2105-2135. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
24
|
Yu M, Fei B, Chu S. Targeting HNRNPA2B1 to overcome chemotherapy resistance in gastric cancer stem cells: Mechanisms and therapeutic potential. J Biol Chem 2025; 301:108234. [PMID: 39870196 PMCID: PMC11999277 DOI: 10.1016/j.jbc.2025.108234] [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: 07/16/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/29/2025] Open
Abstract
Gastric cancer (GC) remains a significant global health challenge, particularly due to the resistance of gastric cancer stem cells (GCSCs) to chemotherapy. This study investigates the role of heterogeneous nuclear ribonucleoprotein A2/B1 (HNRNPA2B1), a member of the heterogeneous nuclear ribonucleoproteins (hnRNPs), in modulating mitochondrial metabolic reprogramming and contributing to chemoresistance in GCSCs. Through extensive analysis of tumor cancer genome atlas (TCGA) and gene expression omnibus (GEO) datasets, HNRNPA2B1 was identified as a key regulator in GCSCs, correlating with poor prognosis and enhanced resistance to chemoresistance. CRISPR-Cas9 mediated knockout of HNRNPA2B1 in GCSCs led to a significant decrease in mitochondrial function, reduced migration, invasion, and sphere formation abilities, and markedly increased apoptosis. These changes were accompanied by a shift in metabolic activity, evidenced by decreased oxygen consumption and increased extracellular acidification. Our results highlight HNRNPA2B1 as a pivotal factor in sustaining the malignant phenotype of GCSCs and present it as a potential therapeutic target to improve chemotherapy efficacy in GC.
Collapse
Affiliation(s)
- Miao Yu
- Department of Gastrointestinal colorectal and anal surgery, The Third Bethune Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Bingyuan Fei
- Department of Gastrointestinal colorectal and anal surgery, The Third Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Songtao Chu
- Department of Forensic Medicine of Basic Medical College, Beihua University, Jilin, Jilin Province, China.
| |
Collapse
|
25
|
Yui K, Imataka G, Ichihashi M. Prostaglandins: Biological Action, Therapeutic Aspects, and Pathophysiology of Autism Spectrum Disorders. Curr Issues Mol Biol 2025; 47:71. [PMID: 39996792 PMCID: PMC11854465 DOI: 10.3390/cimb47020071] [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: 11/07/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Esterified ARA on the inner surface of the cell membrane is hydrolyzed to its free form by phospholipase A2 (PLA2), which is further metabolized by COXs and lipoxygenases (LOXs) and cytochrome P450 (CYP) enzymes. PGs produce detrimental effects due to their proinflammatory properties. The generation of prostaglandin (PG)G2 and PGH2 is triggered by cyclooxygenase (COX) isozymes such as COX-1 and COX-2. Prostaglandin E2 (PGE2) is significantly elevated in ASD. Considerable data indicate that COX enzymes and their metabolites of ARA play important roles in the initiation and development of human neurodevelopmental diseases. The involvement of disrupted COX2/PGE2 signaling in ASD pathology in changing neuronal cell behavior and the expression of ASD-related genes and proteins is due to disrupted COX2/PGE2 signaling. Prostacyclin (PGI2) is synthesized from arachidonic acid by metabolic-pathway-dependent cyclooxygenase (COX) and synthesized in a primary step of ARA transformation (PGG2, PGH2), by degradation of the abovementioned prostaglandins.
Collapse
Affiliation(s)
- Kunio Yui
- Department of Pediatrics, Chiba University, Chiba-Shi 260-8677, Chiba, Japan
| | - George Imataka
- Department of Pediatrics, Dokkyo Medical University, Tochigi 321-0293, Tochigi, Japan;
| | - Mariko Ichihashi
- Department of Orthopedic Surgery, Hyogo Medical University, Nishinomiya 663-8501, Hyogo, Japan
| |
Collapse
|
26
|
Basurco L, Abellanas MA, Purnapatre M, Antonello P, Schwartz M. Chronological versus immunological aging: Immune rejuvenation to arrest cognitive decline. Neuron 2025; 113:140-153. [PMID: 39788084 DOI: 10.1016/j.neuron.2024.12.004] [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: 09/05/2024] [Revised: 10/11/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025]
Abstract
The contemporary understanding that the immune response significantly supports higher brain functions has emphasized the notion that the brain's condition is linked in a complex manner to the state of the immune system. It is therefore not surprising that immunity is a key factor in shaping brain aging. In this perspective article, we propose amending the Latin phrase "mens sana in corpore sano" ("a healthy mind in a healthy body") to "a healthy mind in a healthy immune system." Briefly, we discuss the emerging understanding of the pivotal role of the immune system in supporting lifelong brain maintenance, how the aging of the immune system impacts the brain, and how the potential rejuvenation of the immune system could, in turn, help revitalize brain function, with the ultimate ambitious goal of developing an anti-aging immune therapy.
Collapse
Affiliation(s)
- Leyre Basurco
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Paola Antonello
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
27
|
Groh J, Simons M. White matter aging and its impact on brain function. Neuron 2025; 113:127-139. [PMID: 39541972 DOI: 10.1016/j.neuron.2024.10.019] [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: 08/20/2024] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Aging has a detrimental impact on white matter, resulting in reduced volume, compromised structural integrity of myelinated axons, and an increase in white matter hyperintensities. These changes are closely linked to cognitive decline and neurological disabilities. The deterioration of myelin and its diminished ability to regenerate as we age further contribute to the progression of neurodegenerative disorders. Understanding these changes is crucial for devising effective disease prevention strategies. Here, we will discuss the structural alterations in white matter that occur with aging and examine the cellular and molecular mechanisms driving these aging-related transformations. We highlight how the progressive disruption of white matter may initiate a self-perpetuating cycle of inflammation and neural damage.
Collapse
Affiliation(s)
- Janos Groh
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
28
|
Wang D, Bi X, Zhao L, Xiang S, Xi W, Yang S, Wu W, Chen T, Zheng L, Chi X, Kang Y. Targeting SphK1/S1PR3 axis ameliorates sepsis-induced multiple organ injury via orchestration of macrophage polarization and glycolysis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119877. [PMID: 39549732 DOI: 10.1016/j.bbamcr.2024.119877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
Sepsis is a heterogeneous and imprecise disorder characterized by aberrant response to infection which has been accredited for detrimental impact on immune homeostasis. Recently, macrophage metabolism has been recognized as attractive targets to develop novel immunomodulatory therapy for sepsis research. However, the fine-tuning regulators dictating macrophage functions and the specific mechanisms underlying macrophage metabolic reprogramming remain largely obscure. Sphingosine-1-phosphate (S1P), a metabolic mediator of sphingolipid catabolism, predominantly formed through sphingosine kinase 1 (SphK1) catalyzing, mediates inflammation in sepsis by binding to S1P receptor 3 (S1PR3) expressed in macrophages. Here we demonstrate that SphK1/S1PR3 axis was upregulated in lipopolysaccharide (LPS)-induced macrophages and septic mice lungs, cascading the activation of proglycolytic signaling such as HIF-1α, HK2 and PFKFB3. Targeted inhibition of Sphk1 by PF-543 effectively abrogated upregulated SphK1/S1PR3 axis in vitro and in vivo. In addition, PF-543 significantly suppressed sepsis-related inflammation and multi-organ injury in vivo. Furthermore, PF-543 not only blunted key glycolytic enzymes HIF-1α, HK2, and PFKFB3 in LPS-treated macrophages but also inhibited HK2 and PFKFB3 in septic mice. Silencing or inhibiting SphK1 tempered pro-inflammatory M1 macrophages while boosted anti-inflammatory M2 macrophages. Intriguingly, S1PR3 knockdown proficiently dampened glycolysis-associated markers, retrieved LPS-modulated M1/M2 polarization and attenuated NF-κB p65 activation. In conclusion, our study provides the first evidence that PF-543 orchestrates proportional imbalance of macrophage polarization and the Warburg effect in a SphK1/S1PR3 dependent manner during sepsis, mitigating both hyperinflammation and multi-organ failure, adding a novel puzzle piece to pharmacologically exploitable therapy for sepsis.
Collapse
Affiliation(s)
- Dan Wang
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xinwen Bi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Le Zhao
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shijian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, China
| | - Wenjie Xi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shushu Yang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Weijie Wu
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tufeng Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lei Zheng
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Xinjin Chi
- Surgical Anesthesia Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
29
|
Khan AH, Mulfaul K. Choroidal macrophages in homeostasis, aging and age-related macular degeneration. Exp Eye Res 2025; 250:110159. [PMID: 39577606 DOI: 10.1016/j.exer.2024.110159] [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: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
With increasing age, the optimal functioning of the choroid is essential for efficient removal of waste products formed from photoreceptor renewal. A decline in regulatory elements of the immune system, termed immunosenescence, and the failure of para-inflammation to restore tissue homeostasis can result in the progression of healthy aging to sight-threatening inflammation of the choroid. Macrophages are uniquely situated between the innate and adaptive immune systems, with a high capacity for phagocytosis, recognition of complement components, as well as antigen presentation. In this review, we provide an overview of macrophages and their properties in the healthy choroid and cover the impact of aging, immunosenescence and inflammaging on the function of choroidal macrophages. We will discuss the impact of age on macrophage phenotype and behaviour in the pathophysiology of age-related macular degeneration.
Collapse
Affiliation(s)
- Adnan H Khan
- Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Kelly Mulfaul
- Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology & Visual Sciences, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
30
|
Awogbindin I, ŠimonČiČová E, Vidal V, Ash C, Tremblay ME. Neuroglial responses to bacterial, viral, and fungal neuroinfections. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:213-238. [PMID: 40148046 DOI: 10.1016/b978-0-443-19102-2.00027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Evidence regarding the host's response to peripheral pathogens in humans abound, whereas studies on the pathogenesis of central nervous system-penetrating infections are relatively scarce. However, given the spate of epidemic and pandemic neuroinfections in the 21st century, the field has experienced a renewed interest lately. This chapter discusses a timely and exciting topic on the roles of glial cells, mainly microglia and astrocytes, in neuroinvasive infections. This chapter considered fungal, viral, and bacterial neuroinfections, X-raying their neuroinvasiveness, neurotropism, and neurovirulence before focusing on specific examples notable for each category, including Escherichia coli, Cryptococcus neoformans, and SARS-CoV-2. These infections are renowned worldwide for a high case-fatality rate, leaving many survivors with life-long morbidity and others with a bleak future neurologic prognosis. Importantly, the chapter discusses possible ways microglia and astrocytes are culpable in these infections and provides approaches by which they can be manipulated for therapeutic purposes, identifying viable research gaps in the process. Additionally, it offers a synopsis of ongoing works considering microglial selective targeting to attenuate the pathology, morbidity, and mortality associated with these neuroinfections. Considering that microglia and astrocytes are first responders in the central nervous system, targeting these glial cells could be the game changer in managing existing and emerging neuroinvasive infections.
Collapse
Affiliation(s)
- Ifeoluwa Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada
| | - Eva ŠimonČiČová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Virginie Vidal
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Science and Technology Department, University of Bordeaux, Bordeaux, France
| | - Chantaille Ash
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
31
|
Wang Z, Chen C, Ai J, Gao Y, Wang L, Xia S, Jia Y, Qin Y. The crosstalk between senescence, tumor, and immunity: molecular mechanism and therapeutic opportunities. MedComm (Beijing) 2025; 6:e70048. [PMID: 39811803 PMCID: PMC11731108 DOI: 10.1002/mco2.70048] [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: 06/09/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Cellular senescence is characterized by a stable cell cycle arrest and a hypersecretory, proinflammatory phenotype in response to various stress stimuli. Traditionally, this state has been viewed as a tumor-suppressing mechanism that prevents the proliferation of damaged cells while activating the immune response for their clearance. However, senescence is increasingly recognized as a contributing factor to tumor progression. This dual role necessitates a careful evaluation of the beneficial and detrimental aspects of senescence within the tumor microenvironment (TME). Specifically, senescent cells display a unique senescence-associated secretory phenotype that releases a diverse array of soluble factors affecting the TME. Furthermore, the impact of senescence on tumor-immune interaction is complex and often underappreciated. Senescent immune cells create an immunosuppressive TME favoring tumor progression. In contrast, senescent tumor cells could promote a transition from immune evasion to clearance. Given these intricate dynamics, therapies targeting senescence hold promise for advancing antitumor strategies. This review aims to summarize the dual effects of senescence on tumor progression, explore its influence on tumor-immune interactions, and discuss potential therapeutic strategies, alongside challenges and future directions. Understanding how senescence regulates antitumor immunity, along with new therapeutic interventions, is essential for managing tumor cell senescence and remodeling the immune microenvironment.
Collapse
Affiliation(s)
- Zehua Wang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chen Chen
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiaoyu Ai
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Yaping Gao
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Lei Wang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shurui Xia
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yongxu Jia
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yanru Qin
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
32
|
Jin Z, Jiang C, Cho EB, Bahraminejad S, Han J, Hao J, Liu J, Yu Y, Jiang J. Suppressing the Inflammatory Prostaglandin Signaling after Thrombotic Stroke Ameliorates Ischemic Brain Injury and Facilitates Poststroke Recovery. ACS Pharmacol Transl Sci 2024; 7:4056-4068. [PMID: 39698290 PMCID: PMC11650728 DOI: 10.1021/acsptsci.4c00516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/20/2024]
Abstract
Acute cerebral ischemia is a leading cause of death and disability, particularly among old adults. The narrow therapeutic window and risk of hemorrhagic transformation largely limit patient eligibility for the current treatment. The neuroinflammatory signaling pathway involving the prostaglandin E2 (PGE2) receptor subtype EP2 has now been clarified to contribute to the secondary neurotoxicity following ischemic stroke. We previously demonstrated the feasibility of pharmacologically targeting EP2 for ischemic stroke using an EP2 antagonist in a mouse model of transient middle cerebral artery occlusion. Herein, we evaluated the effects of a second-generation EP2 antagonist with improved potency and selectivity in a mouse model of thrombotic stroke, the most common type of stroke. We found that the EP2 antagonist, when administered hours after an ischemic stroke induced within motor and somatosensory cortices by photoactivation of a light-sensitive dye Rose Bengal, reduced cortical infarction in a dose-dependent manner. EP2 inhibition also improved the poststroke body weight recovery and reduced neurological impairments in locomotor and cognitive functions, revealed by a panel of behavioral tests. These broad benefits support the feasibility of targeting the PGE2/EP2 axis-mediated neuroinflammatory pathway as a novel strategy to alleviate the ischemic brain injury caused by thrombotic occlusion and accelerate poststroke recovery.
Collapse
Affiliation(s)
- Zhen Jin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Chenyao Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Eun Bee Cho
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Sina Bahraminejad
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Juqian Han
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jiukuan Hao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77204, United States
| | - Jiawang Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Medicinal Chemistry Core, Office of Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Ying Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
33
|
Wang X, Hu J, Xie S, Li W, Zhang H, Huang L, Qian Z, Zhao C, Zhang L. Hidden role of microglia during neurodegenerative disorders and neurocritical care: A mitochondrial perspective. Int Immunopharmacol 2024; 142:113024. [PMID: 39217875 DOI: 10.1016/j.intimp.2024.113024] [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: 05/07/2024] [Revised: 08/04/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The incidence of aging-related neurodegenerative disorders and neurocritical care diseases is increasing worldwide. Microglia, the main inflammatory cells in the brain, could be potential viable therapeutic targets for treating neurological diseases. Interestingly, mitochondrial functions, including energy metabolism, mitophagy and transfer, fission and fusion, and mitochondrial DNA expression, also change in activated microglia. Notably, mitochondria play an active and important role in the pathophysiology of neurodegenerative disorders and neurocritical care diseases. This review briefly summarizes the current knowledge on mitochondrial dysfunction in microglia in neurodegenerative disorders and neurocritical care diseases and comprehensively discusses the prospects of the application of neurological injury prevention and treatment targets by mitochondria.
Collapse
Affiliation(s)
- Xinrun Wang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jiyun Hu
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Shucai Xie
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Wenchao Li
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Haisong Zhang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Li Huang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Chunguang Zhao
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - Lina Zhang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
34
|
Mou R, Ma J, Ju X, Wu Y, Chen Q, Li J, Shang T, Chen S, Yang Y, Li Y, Lv K, Chen X, Zhang Q, Liang T, Feng Y, Lu X. Vasopressin drives aberrant myeloid differentiation of hematopoietic stem cells, contributing to depression in mice. Cell Stem Cell 2024; 31:1794-1812.e10. [PMID: 39442524 DOI: 10.1016/j.stem.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/18/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Psychological stress is often linked to depression and can also impact the immune system, illustrating the interconnectedness of mental health and immune function. Hematopoietic stem cells (HSCs) can directly sense neuroendocrine signals in bone marrow and play a fundamental role in the maintenance of immune homeostasis. However, it is unclear how psychological stress impacts HSCs in depression. Here, we report that neuroendocrine factor arginine vasopressin (AVP) promotes myeloid-biased HSC differentiation by activating neutrophils. AVP administration increases neutrophil and Ly6Chi monocyte production by triggering HSCs that rely on intrinsic S100A9 in mice. When stimulated with AVP, neutrophils return to the bone marrow and release interleukin 36G (IL-36G), which interacts with interleukin 1 receptor-like 2 (IL-1RL2) on HSCs to produce neutrophils with high Elane expression that infiltrate the brain and induce neuroinflammation. Together, these findings define HSCs as a relay between psychological stress and myelopoiesis and identify the IL-36G-IL-1RL2 axis as a potential target for depression therapy.
Collapse
Affiliation(s)
- Rong Mou
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Junkai Ma
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Xuan Ju
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310013, Zhejiang, China
| | - Yixin Wu
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
| | - Qiuli Chen
- Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jinglin Li
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Tongyao Shang
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Siying Chen
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Yue Yang
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yue Li
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Kaosheng Lv
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of BioMedical Sciences, Hunan University, Changsha 410028, Hunan, China
| | - Xuequn Chen
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Ye Feng
- Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China.
| | - Xinjiang Lu
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
35
|
Lin CC'J, Tian Y, Tanzi RE, Jorfi M. Approaches for studying neuroimmune interactions in Alzheimer's disease. Trends Immunol 2024; 45:971-986. [PMID: 39537528 PMCID: PMC11624993 DOI: 10.1016/j.it.2024.10.002] [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: 09/09/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Peripheral immune cells play an important role in the pathology of Alzheimer's disease (AD), impacting processes such as amyloid and tau protein aggregation, glial activation, neuronal integrity, and cognitive decline. Here, we examine cutting-edge strategies - encompassing animal and cellular models - used to investigate the roles of peripheral immune cells in AD. Approaches such as antibody-mediated depletion, genetic ablation, and bone marrow chimeras in mouse models have been instrumental in uncovering T, B, and innate immune cell disease-modifying functions. However, challenges such as specificity, off-target effects, and differences between human and mouse immune systems underscore the need for more human-relevant models. Emerging multicellular models replicating critical aspects of human brain tissue and neuroimmune interactions increasingly offer fresh insights into the role of immune cells in AD pathogenesis. Refining these methodologies can deepen our understanding of immune cell contributions to AD and support the development of novel immune-related therapeutic interventions.
Collapse
Affiliation(s)
- Chih-Chung 'Jerry' Lin
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yuyao Tian
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
36
|
Zhao J, Sun Y, Feng Y, Rong J. Brain Specific RagA Overexpression Triggers Depressive-Like Behaviors in Mice via Activating ADORA2A Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404188. [PMID: 39373701 PMCID: PMC11615787 DOI: 10.1002/advs.202404188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/09/2024] [Indexed: 10/08/2024]
Abstract
Neuroinflammation hallmarks the pathology of depression although the etiological complexity has not yet been resolved. Previous studies demonstrate that bacterial lipopolysaccharide induces depressive-like behaviors by activating RagA-mTOR-p70S6K signaling pathway. The current project aims to investigate whether and how brain-specific RagA overexpression triggers depressive-like behaviors in mice. Full-length RagA cDNA is cloned into the mammalian expression vector under the control of brain specific promoter, and subsequently overexpressed in the brain of mouse embryos. Indeed, RagA transgenic mice exhibit depressive-like behaviors and memory impairments. RNA-seq profiling of the prefrontal cortex (PFC) transcriptome highlights adenosine A2a receptor (ADORA2A) as a key differentially expressed gene (DEG). Western blotting confirms that ADORA2A and phospho-p70S6K are markedly elevated in RagA transgenic mice. Behavioral assessments demonstrate that ADORA2A inhibitor istradefylline markedly attenuates depressive-like behaviors. Further metabolomics reveals that N-acetylserotonin and several depression-related metabolites are downregulated while proteomic profiling showed that OLIG1 and other proteins are significantly regulated in RagA transgenic mice. Collectively, RagA overexpression alters the expression patterns of signaling proteins and the metabolism of depression-associated metabolites. RagA may cause depressive-like behaviors in mice via activating p70S6K/ADORA2A signaling pathway. Thus, RagA-p70S6K-ADORA2A signaling pathway may be a target for the development of new antidepressant therapies.
Collapse
Affiliation(s)
- Jia Zhao
- School of Chinese MedicineLi Ka Shing Faculty of MedicineThe University of Hong Kong3 Sassoon Road, PokfulamHong Kong999077P. R. China
- Department of Chinese MedicineThe University of Hong Kong Shenzhen HospitalShenzhen518053P. R. China
| | - Yilu Sun
- School of Chinese MedicineLi Ka Shing Faculty of MedicineThe University of Hong Kong3 Sassoon Road, PokfulamHong Kong999077P. R. China
- Department of Chinese MedicineThe University of Hong Kong Shenzhen HospitalShenzhen518053P. R. China
| | - Yibin Feng
- School of Chinese MedicineLi Ka Shing Faculty of MedicineThe University of Hong Kong3 Sassoon Road, PokfulamHong Kong999077P. R. China
| | - Jianhui Rong
- School of Chinese MedicineLi Ka Shing Faculty of MedicineThe University of Hong Kong3 Sassoon Road, PokfulamHong Kong999077P. R. China
| |
Collapse
|
37
|
Chen SD, Chu CY, Wang CB, Yang Y, Xu ZY, Qu YL, Man Y. Integrated-omics profiling unveils the disparities of host defense to ECM scaffolds during wound healing in aged individuals. Biomaterials 2024; 311:122685. [PMID: 38944969 DOI: 10.1016/j.biomaterials.2024.122685] [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: 02/05/2024] [Revised: 06/11/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Extracellular matrix (ECM) scaffold membranes have exhibited promising potential to better the outcomes of wound healing by creating a regenerative microenvironment around. However, when compared to the application in younger individuals, the performance of the same scaffold membrane in promoting re-epithelialization and collagen deposition was observed dissatisfying in aged mice. To comprehensively explore the mechanisms underlying this age-related disparity, we conducted the integrated analysis, combing single-cell RNA sequencing (scRNA-Seq) with spatial transcriptomics, and elucidated six functionally and spatially distinctive macrophage groups and lymphocytes surrounding the ECM scaffolds. Through intergroup comparative analysis and cell-cell communication, we characterized the dysfunction of Spp1+ macrophages in aged mice impeded the activation of the type Ⅱ immune response, thus inhibiting the repair ability of epidermal cells and fibroblasts around the ECM scaffolds. These findings contribute to a deeper understanding of biomaterial applications in varied physiological contexts, thereby paving the way for the development of precision-based biomaterials tailored specifically for aged individuals in future therapeutic strategies.
Collapse
Affiliation(s)
- Shuai-Dong Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Yu Chu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Bing Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Yang Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhao-Yu Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi-Li Qu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
38
|
Liu Z, Lin Z, Chen Y, Lu M, Hong W, Yu B, Liu G. Lipoteichoic Acid Rescued Age-Related Bone Loss by Enhancing Neuroendocrine and Growth Hormone Secretion Through TLR2/COX2/PGE2 Signalling Pathway. J Cell Mol Med 2024; 28:e70247. [PMID: 39622781 PMCID: PMC11611525 DOI: 10.1111/jcmm.70247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
The phenomenon of brain-bone crosstalk pertains to the intricate interaction and communication pathways between the central nervous system and the skeletal system. Disruption in brain-bone crosstalk, particularly in disorders such as osteoporosis, can result in skeletal irregularities. Consequently, investigating and comprehending this communication network holds paramount importance in the realm of bone disease prevention and management. In this study, we found that Staphylococcus aureus lipoteichoic acid promoted the conversion of arachidonic acid to PGE2 by interacting with TLR2 receptors acting on the surface of microglial cells in the pituitary gland, leading to the upregulation of COX-2 expression. Subsequently, PGE2 bound to the EP4 receptor of growth hormone-secreting cells and activated the intracellular CREB signalling pathway, promoting GH secretion and ameliorating age-related bone loss.
Collapse
Affiliation(s)
- Zixian Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- The Second Hospital and Clinical Medical SchoolLanzhou UniversityLanzhouChina
| | - Zexin Lin
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yingqi Chen
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Mincheng Lu
- Department of Orthopedic, Shenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Weisheng Hong
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Bin Yu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guanqiao Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
39
|
Boonmee A, Benjaskulluecha S, Kueanjinda P, Wongprom B, Pattarakankul T, Sri-Ngern-Ngam K, Umthong S, Takano J, Koseki H, Palaga T. A polycomb group protein EED epigenetically regulates responses in lipopolysaccharide tolerized macrophages. Epigenetics Chromatin 2024; 17:36. [PMID: 39614386 DOI: 10.1186/s13072-024-00562-6] [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: 02/27/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND To avoid exaggerated inflammation, innate immune cells adapt to become hypo-responsive or "tolerance" in response to successive exposure to stimuli, which is a part of innate immune memory. Polycomb repressive complex 2 (PRC2) mediates the transcriptional repression by catalyzing histone H3 lysine 27 trimethylation (H3K27me3) but little is known about its role in lipopolysaccharide (LPS)-induced tolerance in macrophages. RESULT We examined the unexplored roles of EED, a component of the PRC2, in LPS tolerant macrophages. In Eed KO macrophages, significant reduction in H3K27me3 and increased active histone mark, H3K27ac, was observed. Eed KO macrophages exhibited dampened pro-inflammatory cytokine productions (TNF-α and IL-6) while increasing non-tolerizable genes upon LPS tolerance. Pharmacological inhibition of EED also reduced TNF-α and IL-6 during LPS tolerance. Mechanistically, LPS tolerized Eed KO macrophages failed to increase glycolytic activity. RNA-Seq analyses revealed that the hallmarks of hypoxia, TGF-β, and Wnt/β-catenin signaling were enriched in LPS tolerized Eed KO macrophages. Among the upregulated genes, the promoter of Runx3 was found to be associated with EED. Silencing Runx3 in Eed KO macrophages partially rescued the dampened pro-inflammatory response during LPS tolerance. Enrichment of H3K27me3 was decreased in a subset of genes that are upregulated in Eed KO LPS tolerized macrophages, indicating the direct regulatory roles of PRC2 on such genes. Motif enrichment analysis identified the ETS family transcription factor binding sites in the absence of EED in LPS tolerized macrophages. CONCLUSION Our results provided mechanistic insight into how the PRC2 via EED regulates LPS tolerance in macrophages by epigenetically silencing genes that play a crucial role during LPS tolerance such as those of the TGF-β/Runx3 axis.
Collapse
Affiliation(s)
- Atsadang Boonmee
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Immunology and Immune-mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Salisa Benjaskulluecha
- Center of Excellence in Immunology and Immune-mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
- Inter-disciplinary Graduate Program in Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Patipark Kueanjinda
- Center of Excellence in Immunology and Immune-mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Benjawan Wongprom
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Immunology and Immune-mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thitiporn Pattarakankul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Materials and Bio-Interfaces, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kittitach Sri-Ngern-Ngam
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Immunology and Immune-mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supawadee Umthong
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Junichiro Takano
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Immunology and Immune-mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
40
|
Venkataraman A, Kordic I, Li J, Zhang N, Bharadwaj NS, Fang Z, Das S, Coskun AF. Decoding senescence of aging single cells at the nexus of biomaterials, microfluidics, and spatial omics. NPJ AGING 2024; 10:57. [PMID: 39592596 PMCID: PMC11599402 DOI: 10.1038/s41514-024-00178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Aging has profound effects on the body, most notably an increase in the prevalence of several diseases. An important aging hallmark is the presence of senescent cells that no longer multiply nor die off properly. Another characteristic is an altered immune system that fails to properly self-surveil. In this multi-player aging process, cellular senescence induces a change in the secretory phenotype, known as senescence-associated secretory phenotype (SASP), of many cells with the intention of recruiting immune cells to accelerate the clearance of these damaged senescent cells. However, the SASP phenotype results in inducing secondary senescence of nearby cells, resulting in those cells becoming senescent, and improper immune activation resulting in a state of chronic inflammation, called inflammaging, in many diseases. Senescence in immune cells, termed immunosenescence, results in further dysregulation of the immune system. An interdisciplinary approach is needed to physiologically assess aging changes of the immune system at the cellular and tissue level. Thus, the intersection of biomaterials, microfluidics, and spatial omics has great potential to collectively model aging and immunosenescence. Each of these approaches mimics unique aspects of the body undergoes as a part of aging. This perspective highlights the key aspects of how biomaterials provide non-cellular cues to cell aging, microfluidics recapitulate flow-induced and multi-cellular dynamics, and spatial omics analyses dissect the coordination of several biomarkers of senescence as a function of cell interactions in distinct tissue environments. An overview of how senescence and immune dysregulation play a role in organ aging, cancer, wound healing, Alzheimer's, and osteoporosis is included. To illuminate the societal impact of aging, an increasing trend in anti-senescence and anti-aging interventions, including pharmacological interventions, medical procedures, and lifestyle changes is discussed, including further context of senescence.
Collapse
Affiliation(s)
- Abhijeet Venkataraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Ivan Kordic
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - JiaXun Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nicholas Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nivik Sanjay Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Machine Learning Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sandip Das
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
41
|
Sousa NS, Bica M, Brás MF, Sousa AC, Antunes IB, Encarnação IA, Costa TM, Martins IB, Barbosa-Morais NL, Sousa-Victor P, Neves J. The immune landscape of murine skeletal muscle regeneration and aging. Cell Rep 2024; 43:114975. [PMID: 39541212 DOI: 10.1016/j.celrep.2024.114975] [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: 12/04/2023] [Revised: 09/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Age-related alterations in the immune system are starting to emerge as key contributors to impairments found in aged organs. A decline in regenerative capacity is a hallmark of tissue aging; however, the contribution of immune aging to regenerative failure is just starting to be explored. Here, we apply a strategy combining single-cell RNA sequencing with flow cytometry, histological analysis, and functional assays to perform a complete analysis of the immune environment of the aged regenerating skeletal muscle on a time course following injury with single-cell resolution. Our results reveal an unanticipated complexity and functional heterogeneity in immune populations within the skeletal muscle that have been regarded as homogeneous. Furthermore, we uncover a profound remodeling of both myeloid and lymphoid compartments in aging. These discoveries challenge established notions on immune regulation of skeletal muscle regeneration, providing a set of potential targets to improve skeletal muscle health and regenerative capacity in aging.
Collapse
Affiliation(s)
- Neuza S Sousa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Marta Bica
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Margarida F Brás
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Ana C Sousa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Inês B Antunes
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Isabel A Encarnação
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Tiago M Costa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Inês B Martins
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | | | - Pedro Sousa-Victor
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal.
| | - Joana Neves
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal.
| |
Collapse
|
42
|
Liu Z, Lin H, Li X, Xue H, Lu Y, Xu F, Shuai J. The network structural entropy for single-cell RNA sequencing data during skin aging. Brief Bioinform 2024; 26:bbae698. [PMID: 39757115 DOI: 10.1093/bib/bbae698] [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: 10/19/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
Aging is a complex and heterogeneous biological process at cellular, tissue, and individual levels. Despite extensive effort in scientific research, a comprehensive understanding of aging mechanisms remains lacking. This study analyzed aging-related gene networks, using single-cell RNA sequencing data from >15 000 cells. We constructed a gene correlation network, integrating gene expressions into the weights of network edges, and ranked gene importance using a random walk model to generate a gene importance matrix. This unsupervised method improved the clustering performance of cell types. To further quantify the complexity of gene networks during aging, we introduced network structural entropy. The findings of our study reveal that the overall network structural entropy increases in the aged cells compared to the young cells. However, network entropy changes varied greatly within different cell subtypes. Specifically, the network structural entropy among various cell types may increase, remain unchanged, or decrease. This wide range of changes may be closely related to their individual functions, highlighting the cellular heterogeneity and potential key network reconfigurations. Analyzing gene network entropy provides insights into the molecular mechanisms behind aging. This study offers new scientific evidence and theoretical support for understanding the changes in cell functions during aging.
Collapse
Affiliation(s)
- Zhilong Liu
- Department of Physics, Xiamen University, No. 422, Siming South Road, Xiamen, Fujian, 361005, China
| | - Hai Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), No. 999, Jinshi Road, Yongzhong Street, Longwan District, Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou, Zhejiang, 325000, China
| | - Xiang Li
- Department of Physics, Xiamen University, No. 422, Siming South Road, Xiamen, Fujian, 361005, China
| | - Hao Xue
- Department of Computational Biology, Cornell University, 110 Biotechnology Building, Ithaca, 14853 NY, United States
| | - Yuer Lu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), No. 999, Jinshi Road, Yongzhong Street, Longwan District, Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou, Zhejiang, 325000, China
| | - Fei Xu
- Department of Physics, Anhui Normal University, No. 189 Jiuhua South Road, Wuhu, Anhui, 241002, China
| | - Jianwei Shuai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), No. 999, Jinshi Road, Yongzhong Street, Longwan District, Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
43
|
Yu W, Yu Y, Sun S, Lu C, Zhai J, Lei Y, Bai F, Wang R, Chen J. Immune Alterations with Aging: Mechanisms and Intervention Strategies. Nutrients 2024; 16:3830. [PMID: 39599617 PMCID: PMC11597283 DOI: 10.3390/nu16223830] [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: 09/28/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Aging is the result of a complex interplay of physical, environmental, and social factors, leading to an increased prevalence of chronic age-related diseases that burden health and social care systems. As the global population ages, it is crucial to understand the aged immune system, which undergoes declines in both innate and adaptive immunity. This immune decline exacerbates the aging process, creating a feedback loop that accelerates the onset of diseases, including infectious diseases, autoimmune disorders, and cancer. Intervention strategies, including dietary adjustments, pharmacological treatments, and immunomodulatory therapies, represent promising approaches to counteract immunosenescence. These interventions aim to enhance immune function by improving the activity and interactions of aging-affected immune cells, or by modulating inflammatory responses through the suppression of excessive cytokine secretion and inflammatory pathway activation. Such strategies have the potential to restore immune homeostasis and mitigate age-related inflammation, thus reducing the risk of chronic diseases linked to aging. In summary, this review provides insights into the effects and underlying mechanisms of immunosenescence, as well as its potential interventions, with particular emphasis on the relationship between aging, immunity, and nutritional factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (W.Y.)
| |
Collapse
|
44
|
Nunkoo VS, Cristian A, Jurcau A, Diaconu RG, Jurcau MC. The Quest for Eternal Youth: Hallmarks of Aging and Rejuvenating Therapeutic Strategies. Biomedicines 2024; 12:2540. [PMID: 39595108 PMCID: PMC11591597 DOI: 10.3390/biomedicines12112540] [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: 09/30/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The impressive achievements made in the last century in extending the lifespan have led to a significant growth rate of elderly individuals in populations across the world and an exponential increase in the incidence of age-related conditions such as cardiovascular diseases, diabetes mellitus type 2, and neurodegenerative diseases. To date, geroscientists have identified 12 hallmarks of aging (genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, impaired macroautophagy, mitochondrial dysfunction, impaired nutrient sensing, cellular senescence, stem cell exhaustion, defective intercellular communication, chronic inflammation, and gut dysbiosis), intricately linked among each other, which can be targeted with senolytic or senomorphic drugs, as well as with more aggressive approaches such as cell-based therapies. To date, side effects seriously limit the use of these drugs. However, since rejuvenation is a dream of mankind, future research is expected to improve the tolerability of the available drugs and highlight novel strategies. In the meantime, the medical community, healthcare providers, and society should decide when to start these treatments and how to tailor them individually.
Collapse
Affiliation(s)
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
45
|
Forni MF, Pizzurro GA, Krause W, Alexander AF, Bridges K, Xu Y, Justynski O, Gabry A, Camara NOS, Miller-Jensen K, Horsley V. Multiomics reveals age-dependent metabolic reprogramming of macrophages by wound bed niche secreted signals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621159. [PMID: 39553941 PMCID: PMC11565841 DOI: 10.1101/2024.10.30.621159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The cellular metabolism of macrophages depends on tissue niches and can control macrophage inflammatory or resolving phenotypes. Yet, the identity of signals within tissue niches that control macrophage metabolism is not well understood. Here, using single-cell RNA sequencing of macrophages in early mouse wounds, we find that, rather than gene expression of canonical inflammatory or resolving polarization markers, metabolic gene expression defines distinct populations of early wound macrophages. Single-cell secretomics and transcriptomics identify inflammatory and resolving cytokines expressed by early wound macrophages, and we show that these signals drive metabolic inputs and mitochondrial metabolism in an age-dependent manner. We show that aging alters the metabolome of early wound macrophages and rewires their metabolism from mitochondria to glycolysis. We further show that macrophage-derived Chi3l3 and IGF-1 can induce metabolic inputs and mitochondrial mass/metabolism in aged and bone marrow-derived macrophages. Together, these findings reveal that macrophage-derived signals drive the mitochondrial metabolism of macrophages within early wounds in an age-dependent manner and have implications for inflammatory diseases, chronic injuries, and age-related inflammatory diseases. In Brief This study reveals that macrophage subsets in early inflammatory stages of skin wound healing are defined by their metabolic profiles rather than polarization phenotype. Using single-cell secretomics, we establish key macrophage cytokines that comprise the in vivo wound niche and drive mitochondrial-based metabolism. Aging significantly alters macrophage heterogeneity and increases glycolytic metabolism, which can be restored to OxPHOS-based metabolism with young niche cytokines. These findings highlight the importance of the tissue niche in driving macrophage phenotypes, with implications for aging-related impairments in wound healing. Highlights Single cell transcriptional analysis reveals that reveals that metabolic gene expression identifies distinct macrophage populations in early skin wounds.Single-cell secretomic data show that young macrophages contribute to the wound bed niche by secreting molecules such as IGF-1 and Chi3l3.Old wound macrophages display altered metabolomics, elevated glycolytic metabolism and glucose uptake, and reduced lipid uptake and mitochondrial mass/metabolism.Chi3l3 but not IGF-1 secretion is altered in macrophages in an age dependent manner.Chi3l3 can restore mitochondrial mass/metabolism in aged macrophages.
Collapse
|
46
|
Holland C, Dravecz N, Owens L, Benedetto A, Dias I, Gow A, Broughton S. Understanding exogenous factors and biological mechanisms for cognitive frailty: A multidisciplinary scoping review. Ageing Res Rev 2024; 101:102461. [PMID: 39278273 DOI: 10.1016/j.arr.2024.102461] [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: 12/21/2023] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024]
Abstract
Cognitive frailty (CF) is the conjunction of cognitive impairment without dementia and physical frailty. While predictors of each element are well-researched, mechanisms of their co-occurrence have not been integrated, particularly in terms of relationships between social, psychological, and biological factors. This interdisciplinary scoping review set out to categorise a heterogenous multidisciplinary literature to identify potential pathways and mechanisms of CF, and research gaps. Studies were included if they used the definition of CF OR focused on conjunction of cognitive impairment and frailty (by any measure), AND excluded studies on specific disease populations, interventions, epidemiology or prediction of mortality. Searches used Web of Science, PubMed and Science Direct. Search terms included "cognitive frailty" OR (("cognitive decline" OR "cognitive impairment") AND (frail*)), with terms to elicit mechanisms, predictors, causes, pathways and risk factors. To ensure inclusion of animal and cell models, keywords such as "behavioural" or "cognitive decline" or "senescence", were added. 206 papers were included. Descriptive analysis provided high-level categorisation of determinants from social and environmental through psychological to biological. Patterns distinguishing CF from Alzheimer's disease were identified and social and psychological moderators and mediators of underlying biological and physiological changes and of trajectories of CF development were suggested as foci for further research.
Collapse
Affiliation(s)
- Carol Holland
- Division of Health Research, Health Innovation One, Sir John Fisher Drive, Lancaster University, Lancaster LA1 4YW, UK.
| | - Nikolett Dravecz
- Division of Health Research, Health Innovation One, Sir John Fisher Drive, Lancaster University, Lancaster LA1 4YW, UK.
| | - Lauren Owens
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| | - Alexandre Benedetto
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| | - Irundika Dias
- Aston University Medical School, Aston University, Birmingham B4 7ET, UK.
| | - Alan Gow
- Centre for Applied Behavioural Sciences, Department of Psychology, School of Social Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| | - Susan Broughton
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| |
Collapse
|
47
|
Wu L, Lin H, Li S, Huang Y, Sun Y, Shu S, Luo T, Liang T, Lai W, Rao J, Hu Z, Peng H. Macrophage iron dyshomeostasis promotes aging-related renal fibrosis. Aging Cell 2024; 23:e14275. [PMID: 39016438 PMCID: PMC11561705 DOI: 10.1111/acel.14275] [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: 12/22/2023] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Renal aging, marked by the accumulation of senescent cells and chronic low-grade inflammation, leads to renal interstitial fibrosis and impaired function. In this study, we investigate the role of macrophages, a key regulator of inflammation, in renal aging by analyzing kidney single-cell RNA sequencing data of C57BL/6J mice from 8 weeks to 24 months. Our findings elucidate the dynamic changes in the proportion of kidney cell types during renal aging and reveal that increased macrophage infiltration contributes to chronic low-grade inflammation, with these macrophages exhibiting senescence and activation of ferroptosis signaling. CellChat analysis indicates enhanced communications between macrophages and tubular cells during aging. Suppressing ferroptosis alleviates macrophage-mediated tubular partial epithelial-mesenchymal transition in vitro, thereby mitigating the expression of fibrosis-related genes. Using SCENIC analysis, we infer Stat1 as a key age-related transcription factor promoting iron dyshomeostasis and ferroptosis in macrophages by regulating the expression of Pcbp1, an iron chaperone protein that inhibits ferroptosis. Furthermore, through virtual screening and molecular docking from a library of anti-aging compounds, we construct a docking model targeting Pcbp1, which indicates that the natural small molecule compound Rutin can suppress macrophage senescence and ferroptosis by preserving Pcbp1. In summary, our study underscores the crucial role of macrophage iron dyshomeostasis and ferroptosis in renal aging. Our results also suggest Pcbp1 as an intervention target in aging-related renal fibrosis and highlight Rutin as a potential therapeutic agent in mitigating age-related renal chronic low-grade inflammation and fibrosis.
Collapse
Affiliation(s)
- Lingzhi Wu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hongchun Lin
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shaomin Li
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuebo Huang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuxiang Sun
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shuangshuang Shu
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ting Luo
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Tiantian Liang
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Weiyan Lai
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jialing Rao
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhaoyong Hu
- Nephrology Division, Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Hui Peng
- Nephrology Division, Department of Medicine, the Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhouChina
| |
Collapse
|
48
|
Li Z, Chen L, Qu L, Yu W, Liu T, Ning F, Li J, Guo X, Sun F, Sun B, Luo L. Potential implications of natural compounds on aging and metabolic regulation. Ageing Res Rev 2024; 101:102475. [PMID: 39222665 DOI: 10.1016/j.arr.2024.102475] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Aging is generally accompanied by a progressive loss of metabolic homeostasis. Targeting metabolic processes is an attractive strategy for healthy-aging. Numerous natural compounds have demonstrated strong anti-aging effects. This review summarizes recent findings on metabolic pathways involved in aging and explores the anti-aging effects of natural compounds by modulating these pathways. The potential anti-aging effects of natural extracts rich in biologically active compounds are also discussed. Regulating the metabolism of carbohydrates, proteins, lipids, and nicotinamide adenine dinucleotide is an important strategy for delaying aging. Furthermore, phenolic compounds, terpenoids, alkaloids, and nucleotide compounds have shown particularly promising effects on aging, especially with respect to metabolism regulation. Moreover, metabolomics is a valuable tool for uncovering potential targets against aging. Future research should focus on identifying novel natural compounds that regulate human metabolism and should delve deeper into the mechanisms of metabolic regulation using metabolomics methods, aiming to delay aging and extend lifespan.
Collapse
Affiliation(s)
- Zhuozhen Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Lili Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; School of Life Science, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Liangliang Qu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Wenjie Yu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Tao Liu
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Fangjian Ning
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jinwang Li
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Xiali Guo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Fengjie Sun
- Department of Biological Sciences, School of Science and Technology, Georgia Gwinnett College, Lawrenceville, GA 30043, USA
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Liping Luo
- Key Laboratory of Geriatric Nutrition and Health of Ministry of Education, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
49
|
Lyu YX, Fu Q, Wilczok D, Ying K, King A, Antebi A, Vojta A, Stolzing A, Moskalev A, Georgievskaya A, Maier AB, Olsen A, Groth A, Simon AK, Brunet A, Jamil A, Kulaga A, Bhatti A, Yaden B, Pedersen BK, Schumacher B, Djordjevic B, Kennedy B, Chen C, Huang CY, Correll CU, Murphy CT, Ewald CY, Chen D, Valenzano DR, Sołdacki D, Erritzoe D, Meyer D, Sinclair DA, Chini EN, Teeling EC, Morgen E, Verdin E, Vernet E, Pinilla E, Fang EF, Bischof E, Mercken EM, Finger F, Kuipers F, Pun FW, Gyülveszi G, Civiletto G, Zmudze G, Blander G, Pincus HA, McClure J, Kirkland JL, Peyer J, Justice JN, Vijg J, Gruhn JR, McLaughlin J, Mannick J, Passos J, Baur JA, Betts-LaCroix J, Sedivy JM, Speakman JR, Shlain J, von Maltzahn J, Andreasson KI, Moody K, Palikaras K, Fortney K, Niedernhofer LJ, Rasmussen LJ, Veenhoff LM, Melton L, Ferrucci L, Quarta M, Koval M, Marinova M, Hamalainen M, Unfried M, Ringel MS, Filipovic M, Topors M, Mitin N, Roy N, Pintar N, Barzilai N, Binetti P, Singh P, Kohlhaas P, Robbins PD, Rubin P, Fedichev PO, Kamya P, Muñoz-Canoves P, de Cabo R, Faragher RGA, Konrad R, Ripa R, Mansukhani R, et alLyu YX, Fu Q, Wilczok D, Ying K, King A, Antebi A, Vojta A, Stolzing A, Moskalev A, Georgievskaya A, Maier AB, Olsen A, Groth A, Simon AK, Brunet A, Jamil A, Kulaga A, Bhatti A, Yaden B, Pedersen BK, Schumacher B, Djordjevic B, Kennedy B, Chen C, Huang CY, Correll CU, Murphy CT, Ewald CY, Chen D, Valenzano DR, Sołdacki D, Erritzoe D, Meyer D, Sinclair DA, Chini EN, Teeling EC, Morgen E, Verdin E, Vernet E, Pinilla E, Fang EF, Bischof E, Mercken EM, Finger F, Kuipers F, Pun FW, Gyülveszi G, Civiletto G, Zmudze G, Blander G, Pincus HA, McClure J, Kirkland JL, Peyer J, Justice JN, Vijg J, Gruhn JR, McLaughlin J, Mannick J, Passos J, Baur JA, Betts-LaCroix J, Sedivy JM, Speakman JR, Shlain J, von Maltzahn J, Andreasson KI, Moody K, Palikaras K, Fortney K, Niedernhofer LJ, Rasmussen LJ, Veenhoff LM, Melton L, Ferrucci L, Quarta M, Koval M, Marinova M, Hamalainen M, Unfried M, Ringel MS, Filipovic M, Topors M, Mitin N, Roy N, Pintar N, Barzilai N, Binetti P, Singh P, Kohlhaas P, Robbins PD, Rubin P, Fedichev PO, Kamya P, Muñoz-Canoves P, de Cabo R, Faragher RGA, Konrad R, Ripa R, Mansukhani R, Büttner S, Wickström SA, Brunemeier S, Jakimov S, Luo S, Rosenzweig-Lipson S, Tsai SY, Dimmeler S, Rando TA, Peterson TR, Woods T, Wyss-Coray T, Finkel T, Strauss T, Gladyshev VN, Longo VD, Dwaraka VB, Gorbunova V, Acosta-Rodríguez VA, Sorrentino V, Sebastiano V, Li W, Suh Y, Zhavoronkov A, Scheibye-Knudsen M, Bakula D. Longevity biotechnology: bridging AI, biomarkers, geroscience and clinical applications for healthy longevity. Aging (Albany NY) 2024; 16:12955-12976. [PMID: 39418098 PMCID: PMC11552646 DOI: 10.18632/aging.206135] [Show More Authors] [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: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 10/19/2024]
Abstract
The recent unprecedented progress in ageing research and drug discovery brings together fundamental research and clinical applications to advance the goal of promoting healthy longevity in the human population. We, from the gathering at the Aging Research and Drug Discovery Meeting in 2023, summarised the latest developments in healthspan biotechnology, with a particular emphasis on artificial intelligence (AI), biomarkers and clocks, geroscience, and clinical trials and interventions for healthy longevity. Moreover, we provide an overview of academic research and the biotech industry focused on targeting ageing as the root of age-related diseases to combat multimorbidity and extend healthspan. We propose that the integration of generative AI, cutting-edge biological technology, and longevity medicine is essential for extending the productive and healthy human lifespan.
Collapse
Affiliation(s)
- Yu-Xuan Lyu
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Qiang Fu
- Institute of Aging Medicine, College of Pharmacy, Binzhou Medical University, Yantai, China
- Anti-aging Innovation Center, Subei Research Institute at Shanghai Jiaotong University, China
- Shandong Cellogene Pharmaceutics Co. LTD, Yantai, China
| | - Dominika Wilczok
- Duke Kunshan University, Kunshan, Jiangsu, China
- Duke University, Durham, NC, USA
| | - Kejun Ying
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02108, USA
| | - Aaron King
- Foresight Institute, San Francisco, CA 91125, USA
| | - Adam Antebi
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Aleksandar Vojta
- Department of Biology, Division of Molecular Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Alexandra Stolzing
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Alexey Moskalev
- Institute of Biogerontology, Lobachevsky University, Nizhny Novgorod, Russia
| | | | - Andrea B. Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Andrea Olsen
- California Institute of Technology, Pasadena, CA 91125, USA
| | - Anja Groth
- Novo Nordisk Foundation Center for Protein Research (CPR), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Katharina Simon
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- The Kennedy Institute of Rheumatology, Oxford, UK
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Aisyah Jamil
- Insilico Medicine AI Limited, Level 6, Masdar City, Abu Dhabi, UAE
| | - Anton Kulaga
- Systems Biology of Aging Group, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | | | - Benjamin Yaden
- Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University - Purdue University Indianapolis, Indianapolis Indiana 46077, USA
| | | | - Björn Schumacher
- Institute for Genome Stability in Aging and Disease, CECAD Research Center, University and University Hospital of Cologne, Cologne 50931, Germany
| | - Boris Djordjevic
- 199 Biotechnologies Ltd., London, UK
- University College London, London, UK
| | - Brian Kennedy
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chieh Chen
- Molecular, Cellular, And Integrative Physiology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | - Christoph U. Correll
- Zucker School of Medicine at Hofstra/Northwell, NY 10001, USA
- Charité - University Medicine, Berlin, Germany
| | - Coleen T. Murphy
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
- Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
- Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dario Riccardo Valenzano
- Leibniz Institute on Aging, Fritz Lipmann Institute, Friedrich Schiller University, Jena, Germany
| | | | - David Erritzoe
- Centre for Psychedelic Research, Dpt Brain Sciences, Imperial College London, UK
| | - David Meyer
- Institute for Genome Stability in Aging and Disease, CECAD Research Center, University and University Hospital of Cologne, Cologne 50931, Germany
| | - David A. Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 02108, USA
| | - Eduardo Nunes Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, MN 55902, USA
| | - Emma C. Teeling
- School of Biology and Environmental Science, Belfield, Univeristy College Dublin, Dublin 4, Ireland
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Erik Vernet
- Research and Early Development, Maaleov 2760, Denmark
| | | | - Evandro F. Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Evelyne Bischof
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Evi M. Mercken
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Fabian Finger
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen N 2200, Denmark
| | - Folkert Kuipers
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frank W. Pun
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong SAR, China
| | | | | | | | | | - Harold A. Pincus
- Department of Psychiatry, Columbia University, New York, NY 10012, USA
| | | | - James L. Kirkland
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Jan Vijg
- Department of Genetics Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Jennifer R. Gruhn
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Joan Mannick
- Tornado Therapeutics, Cambrian Bio Inc. PipeCo, New York, NY 10012, USA
| | - João Passos
- Department of Physiology and Biomedical Engineering and Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19019, USA
| | | | - John M. Sedivy
- Center on the Biology of Aging, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02860, USA
| | - John R. Speakman
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | | - Julia von Maltzahn
- Faculty of Health Sciences Brandenburg and Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg 01968, Germany
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kelsey Moody
- Ichor Life Sciences, Inc., LaFayette, NY 13084, USA
| | - Konstantinos Palikaras
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55414, USA
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - Liesbeth M. Veenhoff
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lisa Melton
- Nature Biotechnology, Springer Nature, London, UK
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21201, USA
| | - Marco Quarta
- Rubedo Life Sciences, Sunnyvale, CA 94043, USA
- Turn Biotechnologies, Mountain View 94039, CA, USA
- Phaedon Institute, Oakland, CA 94501, USA
| | - Maria Koval
- Institute of Biochemistry of the Romanian Academy, Romania
| | - Maria Marinova
- Fertility and Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Mark Hamalainen
- Longevity Biotech Fellowship, Longevity Acceleration Fund, Vitalism, SF Bay, CA 94101, USA
| | - Maximilian Unfried
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117608, Singapore
| | | | - Milos Filipovic
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V., Dortmund, Germany
| | - Mourad Topors
- Repair Biotechnologies, Inc., Syracuse, NY 13210, USA
| | | | | | | | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10452, USA
| | | | | | | | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology, and Biochemistry, University of Minnesota, Minneapolis, MN 55111, USA
| | | | | | - Petrina Kamya
- Insilico Medicine Canada Inc., Montreal, Quebec H3B 4W8 Canada
| | - Pura Muñoz-Canoves
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA 92121, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging (NIH), Baltimore, Maryland 21201, USA
| | | | | | - Roberto Ripa
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Sabrina Büttner
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm 10691, Sweden
| | - Sara A. Wickström
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | | | | | - Shan Luo
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | | | - Shih-Yin Tsai
- Department of Physiology, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Germany
| | - Thomas A. Rando
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | - Tina Woods
- Collider Heath, London, UK
- Healthy Longevity Champion, National Innovation Centre for Ageing, UK
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15106, USA
| | - Tzipora Strauss
- Sheba Longevity Center, Sheba Medical Center, Tel Hashomer, Israel
- Tel Aviv Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02108, USA
| | - Valter D. Longo
- Longevity Institute, Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90001, USA
| | | | - Vera Gorbunova
- Department of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | - Victoria A. Acosta-Rodríguez
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vincenzo Sorrentino
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA 94301, USA
| | - Wenbin Li
- Department of Neuro-Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University, New York City, NY 10032, USA
| | - Alex Zhavoronkov
- Insilico Medicine AI Limited, Level 6, Masdar City, Abu Dhabi, UAE
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| |
Collapse
|
50
|
Gu X, Fan M, Zhou Y, Zhang Y, Wang L, Gao W, Li T, Wang H, Si N, Wei X, Bian B, Zhao H. Intestinal endogenous metabolites affect neuroinflammation in 5×FAD mice by mediating "gut-brain" axis and the intervention with Chinese Medicine. Alzheimers Res Ther 2024; 16:222. [PMID: 39396997 PMCID: PMC11472645 DOI: 10.1186/s13195-024-01587-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 09/29/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Emerging evidence suggested the association between gut dysbiosis and Alzheimer's disease (AD) progression. However, it remained unclear how the gut microbiome and neuroinflammation in the brain mutually interact or how these interactions affect brain functioning and cognition. Here we hypothesized that "gut-brain" axis mediated by microbial derived metabolites was expected to novel breakthroughs in the fields of AD research and development. METHODS Multiple technologies, such as immunofluorescence, 16s rDNA sequencing, mass spectrometry-based metabolomics (LC-QQQ-MS and GC-MS), were used to reveal potential link between gut microbiota and the metabolism and cognition of the host. RESULTS Microbial depletion induced by the antibiotics mix (ABX) verified that "gut-brain" can transmit information bidirectionally. Short-chain fatty acid-producing (SCFAs-producing) bacteria and amino acid-producing bacteria fluctuated greatly in 5×FAD mice, especially the reduction sharply of the Bifidobacteriaceae and the increase of the Lachnospiraceae family. Concentrations of several Tryptophan-kynurenine intermediates, lactic acid, CD4+ cell, and CD8+ cells were higher in serum of 5×FAD mice, whilst TCA cycle intermediates and Th1/Th2 were lower. In addition, the levels of iso-butyric acid (IBA) in feces, serum, and brain of 5×FAD mice were increased compared with WT-M mice, especially in serum. And IBA in the brain was positively correlated with Aβ and proinflammatory factors. CONCLUSION Together, our finding highlighted that the alternation in gut microbiota affected the effective communication between the "gut-brain" axis in 5×FAD mice by regulating the immune system, carbohydrate, and energy metabolism.
Collapse
Affiliation(s)
- Xinru Gu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- The Neurology Department, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Miaoxuan Fan
- Beijing Drug Package Test Institute, Beijing, 100700, China
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Linna Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wenya Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Hongjie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaolu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|