1
|
Hindman BJ, Olinger CR, Woodroffe RW, Zanaty M, Deifelt Streese C, Zacharias ZR, Houtman JC, Wendt LH, Ten Eyck PP, O'Connell-Moore DJ, Ray EJ, Lee SJ, Waldschmidt DF, Havertape LG, Nguyen LB, Chen PF, Banks MI, Sanders RD, Howard MA. Exploratory randomised trial of tranexamic acid to decrease postoperative delirium in adults undergoing lumbar fusion-a trial stopped early. BJA OPEN 2025; 14:100403. [PMID: 40276619 PMCID: PMC12019205 DOI: 10.1016/j.bjao.2025.100403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/17/2025] [Indexed: 04/26/2025]
Abstract
Background Postoperative delirium may be mediated by systemic inflammation and neuroinflammation. By inhibiting the proinflammatory actions of plasmin, tranexamic acid (TXA) may decrease postoperative delirium. To explore this hypothesis, we modified an ongoing randomised trial of TXA on blood loss, adding measures of delirium, cognition, systemic inflammation, and astrocyte activation. Methods Adults undergoing elective posterior lumbar fusion randomly received intraoperative i.v. TXA (n=43: 10 mg kg-1 loading dose, 2 mg kg-1 h-1 infusion) or placebo (n=40). Blood was collected before surgery and 24 h after surgery (n=32) for biomarkers (cytokines and S100B). Participants had twice daily delirium assessments (n=65). Participants underwent four measures of cognitive function before surgery and during post-discharge follow-up. Results Postoperative blood loss was ∼38% less in the TXA group compared with the placebo group with medians of 128 and 207 ml level-1, respectively, P=0.013. Total blood loss in the TXA and placebo groups did not differ with medians of 305 and 333 ml level-1, respectively, P=0.472. Delirium incidence in the TXA group (7/32=22%) was not significantly less than in the placebo group (11/33=33%); P=0.408, effect size =-0.258 (95% confidence interval -0.744 to 0.229). Conclusions A potential 33% relative decrease in postoperative delirium incidence justifies an adequately powered clinical trial to determine if intraoperative TXA decreases delirium in adults undergoing lumbar fusion. Clinical trial registration NCT04272606.
Collapse
Affiliation(s)
- Bradley J. Hindman
- Department of Anesthesia, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Catherine R. Olinger
- Department of Orthopedics and Rehabilitation, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Royce W. Woodroffe
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mario Zanaty
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | - Zeb R. Zacharias
- Human Immunology Core Laboratory, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Jon C.D. Houtman
- Human Immunology Core Laboratory, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Linder H. Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, USA
| | - Patrick P. Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, USA
| | | | - Emanuel J. Ray
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sarah J. Lee
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Daniel F. Waldschmidt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, USA
| | - Lauren G. Havertape
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, USA
| | - Lanchi B. Nguyen
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, USA
| | - Pei-fu Chen
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Electrical Engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Matthew I. Banks
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Robert D. Sanders
- Central Clinical School and National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Department of Anaesthetics and Institute of Academic Surgery, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Matthew A. Howard
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
2
|
Shaw A, Teng R, Fasina T, Gonzales AS, Wong A, Schweitzer D, Akefe IO. Lipid dysregulation and delirium in older adults: A review of the current evidence and future directions. Brain Res Bull 2025; 224:111299. [PMID: 40086765 DOI: 10.1016/j.brainresbull.2025.111299] [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: 01/07/2025] [Revised: 03/02/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Delirium is a complex medical condition marked by acute episodes of cognitive dysfunction and behavioral disturbances, with a multifaceted etiology and challenging management across various clinical settings. Older adults, particularly in postoperative contexts, are at increased risk of developing delirium. Despite extensive research, a single underlying pathophysiological mechanism for delirium remains elusive. However, emerging evidence suggests a correlation between lipid dysregulation and delirium development in elderly patients, especially in postoperative settings. This connection has led to proposed treatments targeting dyslipidemia and associated neuroinflammatory effects in acute-phase delirium. This review aims to synthesize current literature on the relationship between lipid dysregulation and delirium in older adults, highlighting the need for further research into specific neurolipidome constituents and age-related lipid profile changes, potentially uncovering novel therapeutic strategies for delirium.
Collapse
Affiliation(s)
- AnaLee Shaw
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Rujia Teng
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Toluwani Fasina
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Ana-Sofia Gonzales
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Audrey Wong
- Medical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | | | - Isaac Oluwatobi Akefe
- Academy for Medical Education, The University of Queensland, Herston, QLD 4006, Australia; CDU Menzies School of Medicine, Charles Darwin University, Ellengowan Drive, Darwin, NT 0909, Australia.
| |
Collapse
|
3
|
Kretschmer JR, Tkachenko D, Kümpfel T, Havla J, Engels D, Paul F, Schindler P, Bellmann-Strobl J, Berthele A, Giglhuber K, Zappe C, Klotz L, Revie L, Dawin E, Senel M, Tumani H, Bergh FT, Warnke C, Kraemer M, Walter A, Bayas A, Zettl UK, Lauenstein AS, Yalachkov Y, Etgen T, Kaste M, Luessi F, Gingele S, Passoke S, Weber MS, Sieb JP, Haarmann A, Oschmann P, Rothhammer V, Geis C, Kowarik MC, Kern P, Grothe M, Stephanik H, Angstwurm K, Hoffmann F, Wallwitz U, Wildemann B, Jarius S, Stellmann JP, Pakeerathan T, Schwake C, Ayzenberg I, Kleiter I, Fischer K, Aktas O, Ringelstein M, Häußler V, Trebst C, Hümmert MW. Worse recovery from acute attacks and faster disability accumulation highlights the unmet need for improved treatment in patients with late-onset neuromyelitis optica spectrum disorders (COPTER-LO study). Front Immunol 2025; 16:1575613. [PMID: 40292288 PMCID: PMC12021610 DOI: 10.3389/fimmu.2025.1575613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Objective This study analyzed clinical characteristics, attack recovery and long-term disability accumulation in late-onset (LO ≥ 50 years at onset) versus early-onset (EO < 50 years) NMOSD. Methods This multicenter cohort study included demographic and clinical data from 446 NMOSD patients collected from 35 German Neuromyelitis Optica Study Group (NEMOS) centers. Time to disability milestones was estimated through Kaplan-Meier analysis and Cox proportional hazard regression models adjusted for sex, year of onset, immunotherapy exposure and antibody status. Generalized estimating equations (GEE) were used to compare attack outcomes. Results Of the 446 NMOSD patients analyzed (83.4% female, 85.4% AQP4-IgG-positive, median age at disease onset = 43 years), 153 had a late-onset (34.3%). AQP4-IgG+ prevalence was higher in LO- than in EO-NMOSD (94.1% vs. 80.9%, p<0.001). Optic neuritis at onset was more frequent in EO-NMOSD (27.4% vs. 42.6%, p<0.002), whereas myelitis was more common in LO-NMOSD (58.4% vs. 37.9%, p<0.001). Both groups had similar annualized attack rates (AAR, 0.51 vs. 0.54, p=0.352), but attack recovery was poorer (complete remission in 15.6% vs. 27.4%, p<0.001) and relapse-associated worsening (RAW) was higher in LO-NMOSD (RAW: 3 vs. 0.5, p<0.001). Long-term immunotherapy use was comparable. LO-NMOSD exhibited faster progression to disability endpoints (EDSS 4: HR = 2.64, 95% CI=1.81-3.84). Interpretation LO-NMOSD patients presented more often with myelitis, experienced worse attack outcomes and faster disability accumulation, despite comparable AAR, acute attack treatment and long-term treatment regimens. Accordingly, therapeutic strategies for attack and prophylactic treatment in LO-NMOSD have to be improved.
Collapse
Affiliation(s)
| | - Daria Tkachenko
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, LMU University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, LMU University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Daniel Engels
- Institute of Clinical Neuroimmunology, LMU University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Friedemann Paul
- Department of Neurology, Charite Universitätsmedizin Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Experimental and Clinical Research Center and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Patrick Schindler
- Department of Neurology, Charite Universitätsmedizin Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Experimental and Clinical Research Center and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Judith Bellmann-Strobl
- Department of Neurology, Charite Universitätsmedizin Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Experimental and Clinical Research Center and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Achim Berthele
- Department of Neurology, Technical University Munich, Munich, Germany
| | - Katrin Giglhuber
- Department of Neurology, Technical University Munich, Munich, Germany
| | - Clarissa Zappe
- Department of Neurology, Technical University Munich, Munich, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, Münster, Germany
| | - Lisa Revie
- Department of Neurology, University of Münster, Münster, Germany
| | - Eva Dawin
- Department of Neurology, University of Münster, Münster, Germany
| | - Makbule Senel
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | | | - Clemens Warnke
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Markus Kraemer
- Department of Neurology, Alfried Krupp Hospital Essen, Essen, Germany
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Annette Walter
- Department of Neurology, Herford Hospital, Herford, Germany
| | - Antonios Bayas
- Department of Neurology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Uwe K. Zettl
- Department of Neurology, University of Rostock, Rostock, Germany
| | | | - Yavor Yalachkov
- Department of Neurology, Goethe-University Frankfurt, University Medicine, Frankfurt am Main, Germany
| | - Thorleif Etgen
- Department of Neurology, Klinikum Traunstein, Traunstein, Germany
| | - Matthias Kaste
- Department of Neurology, Nordwest-Krankenhaus Sanderbusch, Sanderbusch, Germany
| | - Felix Luessi
- Department of Neurology, University of Mainz, Mainz, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Sarah Passoke
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Martin S. Weber
- Department of Neurology, University of Göttingen, Göttingen, Germany
| | - Jörn Peter Sieb
- Department of Neurology, Helios Hanseklinikum Stralsund, Stralsund, Germany
| | - Axel Haarmann
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | | | - Veit Rothhammer
- Department of Neurology, FAU Erlangen-Nuremberg, Erlangen, Germany
| | - Christian Geis
- Department of Neurology, University of Jena, Jena, Germany
| | - Markus C. Kowarik
- Department of Neurology & Stroke and Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Peter Kern
- Department of Neurology, Asklepios Expert Clinic Teupitz, Teupitz, Germany
| | - Matthias Grothe
- Department of Neurology, University of Greifswald, Greifswald, Germany
| | - Heike Stephanik
- Department of Neurology, University of Magdeburg, Magdeburg, Germany
| | - Klemens Angstwurm
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Frank Hoffmann
- Department of Neurology, Hospital Martha-Maria, Halle, Germany
| | - Ulrike Wallwitz
- Department of Neurology, Hospital Martha-Maria, Halle, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Jan-Patrick Stellmann
- Institut für Neuroimmunologie und Multiple Sklerose (INIMS), Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
- CEMEREM, APHM, Hôspital de la Timone, Marseille, France
- CRMBM, Aix Marseille Univ, CNRS, Marseille, France
| | - Thivya Pakeerathan
- Department of Neurology, St.Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Carolin Schwake
- Department of Neurology, St.Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Ilya Ayzenberg
- Department of Neurology, St.Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Ingo Kleiter
- Department of Neurology, St.Josef Hospital, Ruhr University Bochum, Bochum, Germany
- Marianne-Strauss-Klinik, Behandlungszentrum für Multiple Sklerose Kranke, Berg, Germany
| | - Katinka Fischer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Herford Hospital, Herford, Germany
| | - Marius Ringelstein
- Department of Neurology, Herford Hospital, Herford, Germany
- Department of Neurology, Center for Neurology and Neuropsychiatry, LVR-Klinikum, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Vivien Häußler
- Institut für Neuroimmunologie und Multiple Sklerose (INIMS), Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Corinna Trebst
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
4
|
Zuo Y, Wang Q, Tian W, Zheng Z, He W, Zhang R, Zhao Q, Miao Y, Yuan Y, Wang J, Zheng H. β-hydroxybutyrylation and O-GlcNAc modifications of STAT1 modulate antiviral defense in aging. Cell Mol Immunol 2025; 22:403-417. [PMID: 39979583 PMCID: PMC11955527 DOI: 10.1038/s41423-025-01266-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/18/2024] [Accepted: 01/30/2025] [Indexed: 02/22/2025] Open
Abstract
Aging changes the protein activity status to affect the body's functions. However, how aging regulates protein posttranslational modifications (PTMs) to modulate the antiviral defense ability of the body remains unclear. Here, we found that aging promotes STAT1 β-hydroxybutyrylation (Kbhb) at Lys592, which inhibits the interaction between STAT1 and type-I interferon (IFN-I) receptor 2 (IFNAR2), thereby attenuating IFN-I-mediated antiviral defense activity. Additionally, we discovered that a small molecule from a plant source, hydroxy camptothecine, can effectively reduce the level of STAT1 Kbhb, thus increasing antiviral defense ability in vivo. Further studies revealed that STAT1 O-GlcNAc modifications at Thr699 block CBP-induced STAT1 Kbhb. Importantly, fructose can improve IFN-I antiviral defense activity by orchestrating STAT1 O-GlcNAc and Kbhb modifications. This study reveals the significance of the switch between STAT1 Kbhb and O-GlcNAc modifications in regulating IFN-I antiviral immunity during aging and provides potential strategies to improve the body's antiviral defense ability in elderly individuals.
Collapse
Affiliation(s)
- Yibo Zuo
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Qin Wang
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Wanying Tian
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Zhijin Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Wei He
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Renxia Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Qian Zhao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Ying Miao
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Yukang Yuan
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Jun Wang
- Department of Intensive Care Medicine, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hui Zheng
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China.
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
- MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, Collaborative Innovation Center of Hematology, School of Medicine, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
5
|
Garcia-Gomara M, Legarra-Marcos N, Serena M, Rojas-de-Miguel E, Espelosin M, Marcilla I, Perez-Mediavilla A, Luquin MR, Lanciego JL, Burrell MA, Cuadrado-Tejedor M, Garcia-Osta A. FKBP51 inhibition ameliorates neurodegeneration and motor dysfunction in the neuromelanin-SNCA mouse model of Parkinson's disease. Mol Ther 2025; 33:895-916. [PMID: 39905728 PMCID: PMC11897814 DOI: 10.1016/j.ymthe.2025.01.049] [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: 08/07/2024] [Revised: 11/16/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of neuromelanin (NM)-containing dopaminergic (DA) neurons in the substantia nigra (SN) pars compacta (SNpc) and the buildup of α-synuclein (α-syn) inclusions, called Lewy bodies. To investigate the roles of NM and α-syn in DA neuron degeneration, we modeled PD by inducing NM accumulation in a humanized α-syn mouse model (Snca-; PAC-Tg(SNCAWT)) via the expression of human tyrosinase in the SN. We found that this mouse strain develops naturally progressive motor dysfunction and dopaminergic neuronal loss in the SN with aging. Upon tyrosinase injection, NM-containing neurons developed p62 and ubiquitin inclusions. Furthermore, the upregulation of genes associated with microglial activation in the midbrain indicated a role of pro-inflammatory factors in neurodegeneration. Midbrain RNA sequencing confirmed the microglial response and identified Fkbp5 as one of the more dysregulated genes. Next, we showed that FKBP51(51 kDa) was significantly upregulated with aging and in PD human brains. Pharmacological treatment with SAFit2, a potent FKBP51 inhibitor, led to a reduction in ubiquitin-positive inclusions, prevention of neurodegeneration in the SNpc, and improved motor function in NM-SNCAWT mice. These results highlight the critical role of FKBP51 in PD and propose SAFit2 as a promising therapeutic candidate for reducing neurodegeneration in PD.
Collapse
Affiliation(s)
- Marta Garcia-Gomara
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Naroa Legarra-Marcos
- Computational Biology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain
| | - Maria Serena
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Elvira Rojas-de-Miguel
- IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Maria Espelosin
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain
| | - Irene Marcilla
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain
| | - Alberto Perez-Mediavilla
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Biochemistry and Genetics Department, School of Sciences, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Maria Rosario Luquin
- IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Neurology, Clínica Universidad de Navarra, University of Navarra, Avenida Pio XII 36, Pamplona, 31008 Navarra, Spain
| | - Jose Luis Lanciego
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain
| | - Maria Angeles Burrell
- IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain
| | - Mar Cuadrado-Tejedor
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Navarra, Spain.
| | - Ana Garcia-Osta
- Gene Therapy for CNS Disorders Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII 55, Pamplona, 31008 Navarra, Spain; IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain.
| |
Collapse
|
6
|
Ma Y, Erb ML, Moore DJ. Aging, cellular senescence and Parkinson's disease. JOURNAL OF PARKINSON'S DISEASE 2025; 15:239-254. [PMID: 39973488 DOI: 10.1177/1877718x251316552] [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: 02/21/2025]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, affecting 1-2% of people over age 65. The risk of developing PD dramatically increases with advanced age, indicating that aging is likely a driving factor in PD neuropathogenesis. Several age-associated biological changes are also hallmarks of PD neuropathology, including mitochondrial dysfunction, oxidative stress, and neuroinflammation. Accumulation of senescent cells is an important feature of aging that contributes to age-related diseases. How age-related cellular senescence affects brain health and whether this phenomenon contributes to neuropathogenesis in PD is not yet fully understood. In this review, we highlight hallmarks of aging, including mitochondrial dysfunction, loss of proteostasis, genomic instability and telomere attrition in relation to well established PD neuropathological pathways. We then discuss the hallmarks of cellular senescence in the context of neuroscience and review studies that directly examine cellular senescence in PD. Studying senescence in PD presents challenges and holds promise for advancing our understanding of disease mechanisms, which could contribute to the development of effective disease-modifying therapeutics. Targeting senescent cells or modulating the senescence-associated secretory phenotype (SASP) in PD requires a comprehensive understanding of the complex relationship between PD pathogenesis and cellular senescence.
Collapse
Affiliation(s)
- Yue Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Madalynn L Erb
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
7
|
Lee CJ, Jang SH, Lim J, Park H, Ahn SH, Park SY, Seo H, Song SJ, Shin JA, Choi C, Gee HY, Choi YH. Exosome-based targeted delivery of NF-κB ameliorates age-related neuroinflammation in the aged mouse brain. Exp Mol Med 2025; 57:235-248. [PMID: 39833561 PMCID: PMC11799301 DOI: 10.1038/s12276-024-01388-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 01/22/2025] Open
Abstract
Neuroinflammation, a significant contributor to various neurodegenerative diseases, is strongly associated with the aging process; however, to date, no efficacious treatments for neuroinflammation have been developed. In aged mouse brains, the number of infiltrating immune cells increases, and the key transcription factor associated with increased chemokine levels is nuclear factor kappa B (NF-κB). Exosomes are potent therapeutics or drug delivery vehicles for various materials, including proteins and regulatory genes, to target cells. In the present study, we evaluated the therapeutic efficacy of exosomes loaded with a nondegradable form of IκB (Exo-srIκB), which inhibits the nuclear translocation of NF-κB to suppress age-related neuroinflammation. Single-cell RNA sequencing revealed that these anti-inflammatory exosomes targeted macrophages and microglia, reducing the expression of inflammation-related genes. Treatment with Exo-srIκB also suppressed the interactions between macrophages/microglia and T and B cells in the aged brain. We demonstrated that Exo-srIκB successfully alleviates neuroinflammation by primarily targeting activated macrophages and partially modulating the functions of age-related interferon-responsive microglia in the brain. Thus, our findings highlight Exo-srIκB as a potential therapeutic agent for treating age-related neuroinflammation.
Collapse
Affiliation(s)
- Chae-Jeong Lee
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Seung Hyun Jang
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jiwoo Lim
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Hyunju Park
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - So-Hee Ahn
- ILIAS Biologics Inc., Daejeon, 34014, Republic of Korea
| | | | - Hyangmi Seo
- ILIAS Biologics Inc., Daejeon, 34014, Republic of Korea
| | - Soo-Jin Song
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Jung-A Shin
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon, 34014, Republic of Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Youn-Hee Choi
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea.
| |
Collapse
|
8
|
Škandík M, Friess L, Vázquez-Cabrera G, Keane L, Grabert K, Cruz De Los Santos M, Posada-Pérez M, Baleviciute A, Cheray M, Joseph B. Age-associated microglial transcriptome leads to diminished immunogenicity and dysregulation of MCT4 and P2RY12/P2RY13 related functions. Cell Death Discov 2025; 11:16. [PMID: 39828750 PMCID: PMC11743796 DOI: 10.1038/s41420-025-02295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The aging process is marked by a time-dependent deterioration in cellular functions, particularly the immune and neural systems. Understanding the phenotype acquisition of microglia, the sentinel immune cells of the brain, is crucial for understanding the nature of age-related neurological diseases. However, the specific phenotype adopted by microglia during aging remains a subject of debate and is contingent on the chosen experimental model. To address these unresolved questions, we employed a novel and highly controlled approach utilizing long-term cultivated BV-2 microglia, exempted from additional external stimuli. Our findings revealed that aged microglial cells, in comparison to their younger counterparts, acquire a distinct gene expression profile, primarily characterized by alterations in microglial immune response. Indeed, pro-inflammatory stimulated aged and young BV-2 microglia exhibited similar transcriptomic profiles, yet the response intensity to the stimulus was markedly muted in the aged microglia. Functional neurotoxic assays confirmed diminished neuronal death in coculture with aged, activated microglia, underscoring a compromised immune response. Furthermore, a subsequent comparative analysis of aged BV-2 microglia with established transcriptomic microglial datasets from aged mice and humans identified 13 overlapping genes, laying the foundation for identifying core microglial aging signature. Particularly noteworthy were SLC16A3 and P2RY13, which consistently exhibited upregulation and downregulation, respectively, across all datasets. Additionally, four other genes-CAPG, LGALS3BP, NRIP1, and P2RY12-were found to share regulatory patterns in response to both aging and extrinsic activation. An in-depth investigation focused on SLC16A3, encoding the high-affinity lactate transporter MCT4, revealed disruptions in extracellular acidification rate and lactate concentration with age. Microglial purine sensing and motility capacities, regulated by P2RY12/P2RY13, displayed age-related alterations. Remarkably, protein analysis in human brain tissue validated the observed upregulation of MCT4 and downregulation of P2RY12 in aged microglia. In conclusion, our study unveils a distinct phenotype in aged microglia characterized by compromised immune responsiveness. Through the integration of in vitro cultured BV-2 microglia with primary microglia datasets, we identify critical molecular determinants of microglial cellular aging confirmed in human-aged brain tissue. This comprehensive approach offers potential insights for understanding and potentially reprogramming aged microglia, with implications for combating age-related neurological disorders.
Collapse
Affiliation(s)
- Martin Škandík
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Friess
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Lily Keane
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kathleen Grabert
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mireia Cruz De Los Santos
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mercedes Posada-Pérez
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China
| | - Austeja Baleviciute
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China.
| |
Collapse
|
9
|
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
|
10
|
Fang S, Jiang M, Jiao J, Zhao H, Liu D, Gao D, Wang T, Yang Z, Yuan H. Unraveling the ROS-Inflammation-Immune Balance: A New Perspective on Aging and Disease. Aging Dis 2025:AD.2024.1253. [PMID: 39812539 DOI: 10.14336/ad.2024.1253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Increased entropy is a common cause of disease and aging. Lifespan entropy is the overall increase in disorder caused by a person over their lifetime. Aging leads to the excessive production of reactive oxygen species (ROS), which damage the antioxidant system and disrupt redox balance. Organ aging causes chronic inflammation, disrupting the balance of proinflammatory and anti-inflammatory factors. Inflammaging, which is a chronic low-grade inflammatory state, is activated by oxidative stress and can lead to immune system senescence. During this process, entropy increases significantly as the body transitions from a state of low order to high disorder. However, the connection among inflammation, aging, and immune system activity is still not fully understood. This review introduces the idea of the ROS-inflammation-immune balance for the first time and suggests that this balance may be connected to aging and the development of age-related diseases. We also explored how the balance of these three factors controls and affects age-related diseases. Moreover, imbalance in the relationship described above disrupts the regular structures of cells and alters their functions, leading to cellular damage and the emergence of a disorganized state marked by increased entropy. Maintaining a low entropy state is crucial for preventing and reversing aging processes. Consequently, we examined the current preclinical evidence for antiaging medications that target this balance. Ultimately, comprehending the intricate relationships between these three factors and the risk of age-related diseases in organisms will aid in the development of clinical interventions that promote long-term health.
Collapse
Affiliation(s)
- Sihang Fang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Mingjun Jiang
- Respiratory Department, Beijing Children's Hospital, Capital Medical University, China National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Juan Jiao
- Department of Clinical Laboratory, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Hongye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dizhi Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Danni Gao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Tenger Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Huiping Yuan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| |
Collapse
|
11
|
Amoriello R, Ballerini C. Immunofluorescence Staining of Murine Spinal Cord Sections to Evaluate Microglial Activation and Astrogliosis. Methods Mol Biol 2025; 2857:159-167. [PMID: 39348064 DOI: 10.1007/978-1-0716-4128-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Microglia and astrocytes are the main components of the central nervous system (CNS). Upon activation, microglia is able to phagocyte cell debris, pathogens, and toxins; astrocytes support neuronal functions, blood-brain barrier (BBB) homeostasis, and neurotransmitter uptake and metabolism. Furthermore, both cell types can produce cytokines and chemokines. Aging impacts microglia and astrocytes by promoting the production of pro-inflammatory cytokines, impairing microglial phagocytosis and motility and astrocyte glutamate uptake. During neurodegenerative and neuroinflammatory diseases, the aging process may be accelerated contributing to the alteration of CNS glial cells functions. Multiple sclerosis (MS) is an autoimmune, demyelinating disease in which immunosenescence can promote the conversion from relapsing-remitting form to progressive disease. The murine model of experimental autoimmune encephalomyelitis (EAE) allows to investigate MS pathogenesis. Furthermore, EAE can be developed as acute or progressive, mimicking different forms of human MS. Microglia and astrocytes report morphological and functional changes during neuroinflammation that can be investigated in different ways. We here present a protocol for the study of glial cell activation in the spinal cord tissue of EAE mice.
Collapse
Affiliation(s)
- Roberta Amoriello
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
12
|
Ding L, Lu Z, Jiang X, Zhang S, Tian X, Wang Q. Obesity-derived macrophages upregulate TNF-α to induce apoptosis in glial cell via the NF-κB/PHLPP1 axis. Int Immunopharmacol 2024; 141:112962. [PMID: 39197294 DOI: 10.1016/j.intimp.2024.112962] [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: 06/03/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
Macrophages in obese adipose tissue have been shown to damage nerve fibers, however, the mechanism underlying how macrophages cause glial cell damage remains unknown. This study aimed to characterize the mechanism by which macrophages induce apoptosis in glial cell during obesity formation in mice by single-nucleus RNA sequencing (snRNA-seq). Cells obtained from paraepididymal adipose tissue in obese mice underwent snRNA-seq. Eighteen different clusters were identified, and 12 cell types were annotated, including glial cells, macrophages, and fibroblasts. There was a negative correlation between the number of glial cells and macrophages in mouse adipose tissue during the formation of obesity. The pro-apoptotic factor PHLPP1 was identified in GO Terms. The interaction between adipose tissue glial cells and macrophages was revealed via in-depth analysis, and the cell-cell communication mechanism between the TNF-α and NF-KB/PHLPP1 axes was perfected. Apoptosis of glial cell by upregulation of TNF-α via obesity-derived macrophages and activation of the NF-κB/PHLPP1 axis. We further revealed how macrophages induce apoptosis in glial cells during obesity formation, as well as different changes in the two cell populations. This study provides valuable resources and foundations for understanding the mechanistic effects of macrophages and glial cells during obesity formation, as well as diseases and potential interventions.
Collapse
Affiliation(s)
- Ling Ding
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Zhimin Lu
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xing Jiang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Sen Zhang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China
| | - Xuewen Tian
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, Jinan, Shandong Province 250102, China.
| |
Collapse
|
13
|
Drake SS, Zaman A, Gianfelice C, Hua EML, Heale K, Afanasiev E, Klement W, Stratton JA, Prat A, Zandee S, Fournier AE. Senolytic treatment diminishes microglia and decreases severity of experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:283. [PMID: 39487537 PMCID: PMC11529445 DOI: 10.1186/s12974-024-03278-2] [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: 05/06/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND The role of senescence in disease contexts is complex, however there is considerable evidence that depletion of senescent cells improves outcomes in a variety of contexts particularly related to aging, cognition, and neurodegeneration. Much research has shown previously that inflammation can promote cellular senescence. Microglia are a central nervous system innate immune cell that undergo senescence with aging and during neurodegeneration. The contribution of senescent microglia to multiple sclerosis, an inflammatory neurodegenerative disease, is not clear, but microglia are strongly implicated in chronic active lesion pathology, tissue injury, and disease progression. Drugs that could specifically eliminate dysregulated microglia in multiple sclerosis are therefore of great interest to the field. RESULTS A single-cell analysis of brain tissue from mice subjected to experimental autoimmune encephalomyelitis (EAE), a mouse model of CNS inflammation that models aspects of multiple sclerosis (MS), identified microglia with a strong transcriptional signature of senescence including the presence of BCL2-family gene transcripts. Microglia expressing Bcl2l1 had higher expression of pro-inflammatory and senescence associated genes than their Bcl2l1 negative counterparts in EAE, suggesting they may exacerbate inflammation. Notably, in human single-nucleus sequencing from MS, BCL2L1 positive microglia were enriched in lesions with active inflammatory pathology, and likewise demonstrated increased expression of immune genes suggesting they may be proinflammatory and contribute to disease processes in chronic active lesions. Employing a small molecule BCL2-family inhibitor, Navitoclax (ABT-263), significantly reduced the presence of microglia and macrophages in the EAE spinal cord, suggesting that these cells can be targeted by senolytic treatment. ABT-263 treatment had a profound effect on EAE mice: decreasing motor symptom severity, improving visual acuity, promoting neuronal survival, and decreasing white matter inflammation. CONCLUSION These results support the hypothesis that microglia and macrophages exhibit transcriptional features of cellular senescence in EAE and MS, and that microglia expressing Bcl2l1 demonstrate a proinflammatory signature that may exacerbate inflammation resulting in negative outcomes in neuroinflammatory disease. Depleting microglia and macrophages using a senolytic results in robust improvement in EAE disease severity, including across measures of neurodegeneration, inflammation, and demyelination, and may therefore represent a novel strategy to address disease progression in multiple sclerosis.
Collapse
Affiliation(s)
- Sienna S Drake
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Aliyah Zaman
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | | | - Elizabeth M-L Hua
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Kali Heale
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Elia Afanasiev
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Wendy Klement
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l', Université de Montréal (CRCHUM), Montreal, Québec, H2X 0A9, Canada
| | - Jo Anne Stratton
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l', Université de Montréal (CRCHUM), Montreal, Québec, H2X 0A9, Canada
| | - Stephanie Zandee
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Alyson E Fournier
- Montréal Neurological Institute, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
14
|
Hindman BJ, Olinger CR, Woodroffe RW, Zanaty M, Streese CD, Zacharias ZR, Houtman JCD, Wendt LH, Eyck PPT, O’Connell-Moore DJ, Ray EJ, Lee SJ, Waldschmidt DF, Havertape LG, Nguyen LB, Chen PF, Banks MI, Sanders RD, Howard MA. Exploratory Randomised Trial of Tranexamic Acid to Decrease Postoperative Delirium in Adults Undergoing Lumbar Fusion: A trial stopped early. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.16.24315638. [PMID: 39484259 PMCID: PMC11527054 DOI: 10.1101/2024.10.16.24315638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background Postoperative delirium may be mediated by perioperative systemic- and neuro-inflammation. By inhibiting the pro-inflammatory actions of plasmin, tranexamic acid (TXA) may decrease postoperative delirium. To explore this hypothesis, we modified an ongoing randomised trial of TXA, adding measures of postoperative delirium, cognitive function, systemic cytokines, and astrocyte activation. Methods Adults undergoing elective posterior lumbar fusion randomly received intraoperative intravenous TXA (n=43: 10 mg kg-1 loading dose, 2 mg kg-1 h-1 infusion) or Placebo (n=40). Blood was collected pre- and at 24 h post-operatively (n=32) for biomarkers of systemic inflammation (cytokines) and astrocyte activation (S100B). Participants had twice daily delirium assessments using the 3-minute diagnostic interview for Confusion Assessment Method (n=65). Participants underwent 4 measures of cognitive function preoperatively and during post-discharge follow-up. Results Delirium incidence in the TXA group (7/32=22%) was not significantly less than in the Placebo group (11/33=33%); P=0.408, absolute difference=11%, relative difference=33%, effect size = -0.258 (95% CI -0.744 to 0.229). In the Placebo group (n=16), delirium severity was associated with the number of instrumented vertebral levels (P=0.001) and with postoperative interleukin -8 and -10 concentrations (P=0.00008 and P=0.005, respectively) and these associations were not significantly modified by TXA. In the Placebo group, delirium severity was associated with S100B concentration (P=0.0009) and the strength of the association was decreased by TXA (P=0.002). Conclusions A potential 33% relative decrease in postoperative delirium incidence justifies an adequately powered clinical trial to determine if intraoperative TXA decreases delirium in adults undergoing lumbar fusion.
Collapse
Affiliation(s)
- Bradley J. Hindman
- Department of Anesthesia, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Catherine R. Olinger
- Department of Orthopedics and Rehabilitation, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Royce W. Woodroffe
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Mario Zanaty
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | | | - Zeb R. Zacharias
- Human Immunology Core Laboratory, University of Iowa, Iowa City, Iowa, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Jon C. D. Houtman
- Human Immunology Core Laboratory, University of Iowa, Iowa City, Iowa, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Linder H. Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Patrick P. Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Debra J. O’Connell-Moore
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Emanuel J. Ray
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Sarah J. Lee
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Daniel F. Waldschmidt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Lauren G. Havertape
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Lanchi B. Nguyen
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Pei-fu Chen
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Electrical Engineering, Yuan Ze University, Taoyuan City, 320315, Taiwan
| | - Matthew I. Banks
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Robert D. Sanders
- Central Clinical School and National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, Australia
- Department of Anaesthetics and Institute of Academic Surgery, Royal Prince Alfred Hospital, Sydney, Australia
| | - Matthew A. Howard
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
15
|
Cao S, Wang S, Luo H, Guo J, Xuan L, Sun L. The effect of macrophage-cardiomyocyte interactions on cardiovascular diseases and development of potential drugs. Mol Biol Rep 2024; 51:1056. [PMID: 39417949 DOI: 10.1007/s11033-024-09944-1] [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: 04/01/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
The interaction between macrophages and cardiomyocytes plays an important role not only in maintaining cardiac homeostasis, but also in the development of many cardiovascular diseases (CVDs), such as myocardial infarction (MI) and heart failure (HF). In addition to supporting cardiomyocytes, macrophages and cardiomyocytes have a close and complex relationship. By studying their cross-talk, we can better understand novel mechanisms and target pathogenic mechanisms, and improve the treatment of CVDs. We review macrophage-cardiomyocyte communication through connexin 43 (Cx43)-containing gap junctions (GJs) directly, secreted protein factors indirectly, and discuss the implications of these interactions in cardiac homeostasis and the development of various CVDs, including MI, HF, arrhythmia, cardiac fibrosis and myocarditis. In this section, we review various drugs that work by modulating cytokines or other proteins to reduce inflammation in CVDs. The clinical findings from targeting inflammation in CVDs are also discussed. Additionally, we examine the challenges and opportunities for improving our understanding of macrophage-cardiomyocyte coupling as it relates to pathophysiological disease processes, extending our research scope, and helping identify new molecular targets and improve the effectiveness of existing therapies.
Collapse
Affiliation(s)
- Shoupeng Cao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Shengjie Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Huishan Luo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Jianjun Guo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Lina Xuan
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medicial University, Harbin, 157 Baojian Road, Nangang District, 150081, heilongjiang, China.
| | - Lihua Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University (Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medicial University, Harbin, 157 Baojian Road, Nangang District, 150081, heilongjiang, China.
| |
Collapse
|
16
|
Krsek A, Ostojic L, Zivalj D, Baticic L. Navigating the Neuroimmunomodulation Frontier: Pioneering Approaches and Promising Horizons-A Comprehensive Review. Int J Mol Sci 2024; 25:9695. [PMID: 39273641 PMCID: PMC11396210 DOI: 10.3390/ijms25179695] [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: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The research in neuroimmunomodulation aims to shed light on the complex relationships that exist between the immune and neurological systems and how they affect the human body. This multidisciplinary field focuses on the way immune responses are influenced by brain activity and how neural function is impacted by immunological signaling. This provides important insights into a range of medical disorders. Targeting both brain and immunological pathways, neuroimmunomodulatory approaches are used in clinical pain management to address chronic pain. Pharmacological therapies aim to modulate neuroimmune interactions and reduce inflammation. Furthermore, bioelectronic techniques like vagus nerve stimulation offer non-invasive control of these systems, while neuromodulation techniques like transcranial magnetic stimulation modify immunological and neuronal responses to reduce pain. Within the context of aging, neuroimmunomodulation analyzes the ways in which immunological and neurological alterations brought on by aging contribute to cognitive decline and neurodegenerative illnesses. Restoring neuroimmune homeostasis through strategies shows promise in reducing age-related cognitive decline. Research into mood disorders focuses on how immunological dysregulation relates to illnesses including anxiety and depression. Immune system fluctuations are increasingly recognized for their impact on brain function, leading to novel treatments that target these interactions. This review emphasizes how interdisciplinary cooperation and continuous research are necessary to better understand the complex relationship between the neurological and immune systems.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Leona Ostojic
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Dorotea Zivalj
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
17
|
Zhao Y, Si S, Ren Y, Wu X, Zhang Z, Tian Y, Li J, Li Y, Hou M, Yao X, Xu Z, Jiang R, Kang X, Gong Y, Li Q, Tian Y. Marine red yeast supplementation improves laying performance by regulating small intestinal homeostasis in aging chickens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:177-190. [PMID: 39263442 PMCID: PMC11388669 DOI: 10.1016/j.aninu.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/25/2024] [Accepted: 04/03/2024] [Indexed: 09/13/2024]
Abstract
Recent studies have shown that age-related aging evolution is accompanied by imbalances in intestinal homeostasis. Marine red yeast (MRY) is a functional probiotic that has been shown to have antioxidant, immune and other properties. Therefore, we chose 900 healthy Hy-Line Brown hens at 433 d old as the research subjects and evaluated the correlation between intestinal health, laying performance, and egg quality in aged hens through the supplementation of MRY. These laying hens were assigned into 5 groups and received diet supplementation with 0%, 0.5%, 1.0%, 1.5%, and 2% MRY for 12 weeks. The results showed that MRY supplementation increased egg production rate, average egg weight, and egg quality, and decreased feed conversion ratio and daily feed intake (P < 0.05). The MRY supplement improved antioxidant indicators such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), stimulated villus height, and increased the villus height to crypt depth ratio (V/C ratio) in the intestine (P < 0.05). It also regulated the expression of intestinal inflammatory factors (transforming growth factor-β [TGF-β], interleukin [IL]-1β, IL-8, tumor necrosis factor-α [TNF-α]) while increasing serum immunoglobulin G (IgG) levels (P < 0.05). Furthermore, MRY supplementation upregulated the mRNA expression of tight junction proteins (occludin and zonula occludens-1 [ZO-1]), anti-apoptotic gene (Bcl-2), and autophagy-related proteins (beclin-1 and light chain 3I [LC3I]) in the intestine (P < 0.05). The MRY supplement also led to an increase in the concentration of short-chain fatty acids in the cecum, and the relative abundance of the phylum Bacteroidetes, and genera Bacteroides and Rikenellaceae_RC9_gut_group. The LEfSe analysis revealed an enrichment of Sutterella and Akkermansia muciniphila. In conclusion, the results of this experiment indicated that the additional supplementation of MRY can improve the production performance of laying hens and may contribute to the restoration and balance of intestinal homeostasis, which supports the application potential of MRY as a green and efficient feed additive for improving the laying performance in chickens.
Collapse
Affiliation(s)
- Yudian Zhao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Sujin Si
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yangguang Ren
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xing Wu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Zihao Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yixiang Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Jing Li
- AB Vista, Marlborough SN8 4AN, UK
| | - Yijie Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Meng Hou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xueyang Yao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhaoheng Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yujie Gong
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Qiang Li
- Henan College of Animal Husbandry and Economics, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| |
Collapse
|
18
|
Clark D, Brazina S, Miclau T, Park S, Hsieh CL, Nakamura M, Marcucio R. Age-related changes to macrophage subpopulations and TREM2 dysregulation characterize attenuated fracture healing in old mice. Aging Cell 2024; 23:e14212. [PMID: 38825965 PMCID: PMC11488338 DOI: 10.1111/acel.14212] [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: 02/06/2024] [Revised: 04/11/2024] [Accepted: 05/10/2024] [Indexed: 06/04/2024] Open
Abstract
Fracture healing complications increase with age, with higher rates of delayed unions and nonunions and an associated increase in morbidity and mortality in older adults. Macrophages have a dynamic role in fracture healing, and we have previously demonstrated that age-related changes in macrophages are associated with attenuated fracture repair in old mice. Here, we provide a single cell characterization of the immune cells involved in the early phase of fracture healing. We show that there were multiple transcriptionally distinct macrophage subpopulations present simultaneously within the healing tissue. Fracture healing was attenuated in old mice compared to young, and macrophages from the fracture callus of old mice demonstrated a pro-inflammatory phenotype compared to young. Interestingly, Trem2 expression was decreased in old macrophages compared to young. Young mice lacking Trem2 demonstrated attenuated fracture healing and inflammatory dysregulation similar to old mice. Trem2 dysregulation has previously been implicated in other age-related diseases, but its role in fracture healing is unknown. This work provides a robust characterization of the macrophage subpopulations involved in fracture healing, and further reveals the important role of Trem2 in fracture healing and may be a potential driver of age-related inflammatory dysregulation. Future work may further examine macrophages and Trem2 as potential therapeutic targets for management of fracture repair in older adults.
Collapse
Affiliation(s)
- Daniel Clark
- Center for Craniofacial RegenerationUniversity of Pittsburgh School of Dental MedicinePittsburghPennsylvaniaUSA
- Department of Periodontics and Preventive DentistryUniversity of Pittsburgh School of Dental MedicinePittsburghPennsylvaniaUSA
| | - Sloane Brazina
- Department of Obstetrics and GynecologyUniversity of California DavisSacramentoCaliforniaUSA
| | - Ted Miclau
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, School of MedicineUniversity of California San Francisco, Zuckerberg San Francisco General HospitalSan FranciscoCaliforniaUSA
| | - Sangmin Park
- Center for Craniofacial RegenerationUniversity of Pittsburgh School of Dental MedicinePittsburghPennsylvaniaUSA
| | - Christine L. Hsieh
- Division of Rheumatology, Department of MedicineUniversity of California San Francisco, San Francisco VA Health Care SystemSan FranciscoCaliforniaUSA
| | - Mary Nakamura
- Division of Rheumatology, Department of MedicineUniversity of California San Francisco, San Francisco VA Health Care SystemSan FranciscoCaliforniaUSA
| | - Ralph Marcucio
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, School of MedicineUniversity of California San Francisco, Zuckerberg San Francisco General HospitalSan FranciscoCaliforniaUSA
| |
Collapse
|
19
|
Mi B, Xiong Y, Knoedler S, Alfertshofer M, Panayi AC, Wang H, Lin S, Li G, Liu G. Ageing-related bone and immunity changes: insights into the complex interplay between the skeleton and the immune system. Bone Res 2024; 12:42. [PMID: 39103328 PMCID: PMC11300832 DOI: 10.1038/s41413-024-00346-4] [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: 11/16/2023] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
Ageing as a natural irreversible process inherently results in the functional deterioration of numerous organ systems and tissues, including the skeletal and immune systems. Recent studies have elucidated the intricate bidirectional interactions between these two systems. In this review, we provide a comprehensive synthesis of molecular mechanisms of cell ageing. We further discuss how age-related skeletal changes influence the immune system and the consequent impact of immune system alterations on the skeletal system. Finally, we highlight the clinical implications of these findings and propose potential strategies to promote healthy ageing and reduce pathologic deterioration of both the skeletal and immune systems.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Michael Alfertshofer
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig - Maximilian University Munich, Munich, Germany
| | - Adriana C Panayi
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Hand-, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Haixing Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
20
|
Xu Y, Bai L, Yang X, Huang J, Wang J, Wu X, Shi J. Recent advances in anti-inflammation via AMPK activation. Heliyon 2024; 10:e33670. [PMID: 39040381 PMCID: PMC11261115 DOI: 10.1016/j.heliyon.2024.e33670] [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: 10/09/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Inflammation is a complex physiological phenomenon, which is the body's defensive response, but abnormal inflammation can have adverse effects, and many diseases are related to the inflammatory response. AMPK, as a key sensor of cellular energy status, plays a crucial role in regulating cellular energy homeostasis and glycolipid metabolism. In recent years, the anti-inflammation effect of AMPK and related signalling cascade has begun to enter everyone's field of vision - not least the impact on metabolic diseases. A great number of studies have shown that anti-inflammatory drugs work through AMPK and related pathways. Herein, this article summarises recent advances in compounds that show anti-inflammatory effects by activating AMPK and attempts to comment on them.
Collapse
Affiliation(s)
- Yihua Xu
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xinwei Yang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Jianli Huang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jie Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xianbo Wu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Liu J, Kong G, Lu C, Wang J, Li W, Lv Z, Tong J, Liu Y, Xiong W, Li H, Fan J. IPSC-NSCs-derived exosomal let-7b-5p improves motor function after spinal cord Injury by modulating microglial/macrophage pyroptosis. J Nanobiotechnology 2024; 22:403. [PMID: 38982427 PMCID: PMC11232148 DOI: 10.1186/s12951-024-02697-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Following spinal cord injury (SCI), the inflammatory storm initiated by microglia/macrophages poses a significant impediment to the recovery process. Exosomes play a crucial role in the transport of miRNAs, facilitating essential cellular communication through the transfer of genetic material. However, the miRNAs from iPSC-NSCs-Exos and their potential mechanisms leading to repair after SCI remain unclear. This study aims to explore the role of iPSC-NSCs-Exos in microglia/macrophage pyroptosis and reveal their potential mechanisms. METHODS iPSC-NSCs-Exos were characterized and identified using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot. A mouse SCI model and a series of in vivo and in vitro experiments were conducted to investigate the therapeutic effects of iPSC-NSCs-Exos. Subsequently, miRNA microarray analysis and rescue experiments were performed to confirm the role of miRNAs in iPSC-NSCs-Exos in SCI. Mechanistic studies were carried out using Western blot, luciferase activity assays, and RNA-ChIP. RESULTS Our findings revealed that iPSC-NSCs-derived exosomes inhibited microglia/macrophage pyroptosis at 7 days post-SCI, maintaining myelin integrity and promoting axonal growth, ultimately improving mice motor function. The miRNA microarray showed let-7b-5p to be highly enriched in iPSC-NSCs-Exos, and LRIG3 was identified as the target gene of let-7b-5p. Through a series of rescue experiments, we uncovered the connection between iPSC-NSCs and microglia/macrophages, revealing a novel target for treating SCI. CONCLUSION In conclusion, we discovered that iPSC-NSCs-derived exosomes can package and deliver let-7b-5p, regulating the expression of LRIG3 to ameliorate microglia/macrophage pyroptosis and enhance motor function in mice after SCI. This highlights the potential of combined therapy with iPSC-NSCs-Exos and let-7b-5p in promoting functional recovery and limiting inflammation following SCI.
Collapse
Affiliation(s)
- Jie Liu
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China
| | - Guang Kong
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chenlin Lu
- Department of Clinical Research Center, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
| | - Juan Wang
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenbo Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China
| | - Zhengming Lv
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
| | - Jian Tong
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
| | - Yuan Liu
- Songjiang Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wu Xiong
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China.
| | - Haijun Li
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China.
| | - Jin Fan
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China.
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China.
| |
Collapse
|
22
|
Rosmus DD, Koch J, Hausmann A, Chiot A, Arnhold F, Masuda T, Kierdorf K, Hansen SM, Kuhrt H, Fröba J, Wolf J, Boneva S, Gericke M, Ajami B, Prinz M, Lange C, Wieghofer P. Redefining the ontogeny of hyalocytes as yolk sac-derived tissue-resident macrophages of the vitreous body. J Neuroinflammation 2024; 21:168. [PMID: 38961498 PMCID: PMC11223341 DOI: 10.1186/s12974-024-03110-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/22/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The eye is a highly specialized sensory organ which encompasses the retina as a part of the central nervous system, but also non-neural compartments such as the transparent vitreous body ensuring stability of the eye globe and a clear optical axis. Hyalocytes are the tissue-resident macrophages of the vitreous body and are considered to play pivotal roles in health and diseases of the vitreoretinal interface, such as proliferative vitreoretinopathy or diabetic retinopathy. However, in contrast to other ocular macrophages, their embryonic origin as well as the extent to which these myeloid cells might be replenished by circulating monocytes remains elusive. RESULTS In this study, we combine transgenic reporter mice, embryonic and adult fate mapping approaches as well as parabiosis experiments with multicolor immunofluorescence labeling and confocal laser-scanning microscopy to comprehensively characterize the murine hyalocyte population throughout development and in adulthood. We found that murine hyalocytes express numerous well-known myeloid cell markers, but concomitantly display a distinct immunophenotype that sets them apart from retinal microglia. Embryonic pulse labeling revealed a yolk sac-derived origin of murine hyalocytes, whose precursors seed the developing eye prenatally. Finally, postnatal labeling and parabiosis established the longevity of hyalocytes which rely on Colony Stimulating Factor 1 Receptor (CSF1R) signaling for their maintenance, independent of blood-derived monocytes. CONCLUSION Our study identifies hyalocytes as long-living progeny of the yolk sac hematopoiesis and highlights their role as integral members of the innate immune system of the eye. As a consequence of their longevity, immunosenescence processes may culminate in hyalocyte dysfunction, thereby contributing to the development of vitreoretinal diseases. Therefore, myeloid cell-targeted therapies that convey their effects through the modification of hyalocyte properties may represent an interesting approach to alleviate the burden imposed by diseases of the vitreoretinal interface.
Collapse
Affiliation(s)
- Dennis-Dominik Rosmus
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Augsburg University, Universitätsstrasse 2, 86159, Augsburg, Germany
| | - Jana Koch
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Augsburg University, Universitätsstrasse 2, 86159, Augsburg, Germany
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Annika Hausmann
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Aude Chiot
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Franz Arnhold
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Division of Molecular Neuroimmunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Katrin Kierdorf
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies, University of Freiburg, 79106, Freiburg, Germany
- Centre for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Stefanie Marie Hansen
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Heidrun Kuhrt
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Janine Fröba
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Julian Wolf
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, 94304, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Bahareh Ajami
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Marco Prinz
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79106, Freiburg, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Ophtha Lab, Department of Ophthalmology, St. Franziskus Hospital, 48145, Münster, Germany
| | - Peter Wieghofer
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany.
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Augsburg University, Universitätsstrasse 2, 86159, Augsburg, Germany.
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.
| |
Collapse
|
23
|
de Seze J, Dive D, Ayrignac X, Castelnovo G, Payet M, Rayah A, Gobbi C, Vermersch P, Zecca C. Narrative Review on the Use of Cladribine Tablets as Exit Therapy for Stable Elderly Patients with Multiple Sclerosis. Neurol Ther 2024; 13:519-533. [PMID: 38587749 PMCID: PMC11136913 DOI: 10.1007/s40120-024-00603-y] [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: 02/13/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2024] Open
Abstract
The number of ageing people with relapsing multiple sclerosis (RMS) is increasing. The efficacy of disease-modifying therapies (DMTs) for RMS declines with age. Also, older persons with MS may be more susceptible to infections, hospitalisations and malignancy. Aging people with MS have higher rates of comorbidities versus aged-matched controls, increasing the individual risk of disability. We review the therapeutic properties of cladribine tablets (CladT) in ageing people with RMS, with regard to their utility for allowing these individuals to cease continuous administration of a DMT (i.e. to act as an "exit therapy"). CladT is thought to be an immune reconstitution therapy, in that two short courses of oral treatment 1 year apart provide suppression of MS disease activity in responders that far outlasts the duration of treatment and post-treatment reductions in lymphocyte counts. Post hoc analyses, long-term follow-up of populations with RMS in randomised trials, and real-world evidence suggest that the efficacy of CladT is probably independent of age, although more data in the elderly are still needed. No clear adverse signals for lymphopenia or other adverse safety signals have emerged with increasing age, although immunosenescence in the setting of age-related "inflammaging" may predispose elderly patients to a higher risk of infections. Updating vaccination status is recommended, especially against pneumococci and herpes zoster for older patients, to minimise the risk of these infections. CladT may be a useful alternative treatment for ageing people with MS who often bear a burden of multiple comorbidities and polypharmacy and who are more exposed to the adverse effects of continuous immunosuppressive therapy.
Collapse
Affiliation(s)
- Jerome de Seze
- Department of Neurology, Strasbourg University Hospital, Strasbourg, France.
| | - Dominique Dive
- Department of Neurology, Liège University Hospital, Liège, Belgium
| | - Xavier Ayrignac
- Department of Neurology, University of Montpellier, INM, INSERM, Montpellier University Hospital, Montpellier, France
| | - Giovanni Castelnovo
- Department of Neurology, Nîmes University Hospital, Hopital Caremeau, Nîmes, France
| | - Marianne Payet
- Merck Santé S.A.S., an Affiliate of Merck KGaA, Lyon, France
| | - Amel Rayah
- Merck Santé S.A.S., an Affiliate of Merck KGaA, Lyon, France
| | - Claudio Gobbi
- Multiple Sclerosis Center, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Patrick Vermersch
- University of Lille, INSERM U1172 LilNCog, CHU Lille, FHU Precise, Lille, France
| | - Chiara Zecca
- Multiple Sclerosis Center, Neurocenter of Southern Switzerland, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
24
|
Li C, Stebbins RC, Noppert GA, Carney CX, Liu C, Sapp ARM, Watson EJ, Aiello AE. Peripheral immune function and Alzheimer's disease: a living systematic review and critical appraisal. Mol Psychiatry 2024; 29:1895-1905. [PMID: 38102484 PMCID: PMC11483233 DOI: 10.1038/s41380-023-02355-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND A growing body of literature examines the relationship between peripheral immune function and Alzheimer's Disease (AD) in human populations. Our living systematic review summarizes the characteristics and findings of these studies, appraises their quality, and formulates recommendations for future research. METHODS We searched the electronic databases PubMed, PsycINFO, and Web of Science, and reviewed references of previous reviews and meta-analyses to identify human studies examining the relationship between any peripheral immune biomarkers and AD up to September 7th, 2023. We examined patterns of reported statistical associations (positive, negative, and null) between each biomarker and AD across studies. Evidence for each biomarker was categorized into four groups based on the proportion of studies reporting different associations: corroborating a positive association with AD, a negative association, a null association, and presenting contradictory findings. A modified Newcastle-Ottawa scale (NOS) was employed to assess the quality of the included studies. FINDINGS In total, 286 studies were included in this review. The majority were cross-sectional (n = 245, 85.7%) and hospital-based (n = 248, 86.7%), examining relationships between 187 different peripheral immune biomarkers and AD. Cytokines were the most frequently studied group of peripheral immune biomarkers. Evidence supported a positive association with AD for six biomarkers, including IL-6, IL-1β, IFN-γ, ACT, IL-18, and IL-12, and a negative association for two biomarkers, including lymphocytes and IL-6R. Only a small proportion of included studies (n = 22, 7.7%) were deemed to be of high quality based on quality assessment. INTERPRETATION Existing research on peripheral immune function and AD exhibits substantial methodological variations and limitations, with a notable lack of longitudinal, population-based studies investigating a broad range of biomarkers with prospective AD outcomes. The extent and manner in which peripheral immune function can contribute to AD pathophysiology remain open questions. Given the biomarkers that we identified to be associated with AD, we posit that targeting peripheral immune dysregulation may present a promising intervention point to reduce the burden of AD.
Collapse
Affiliation(s)
- Chihua Li
- Social Environment and Health Program, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology, School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| | - Rebecca C Stebbins
- Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York City, NY, USA
| | - Grace A Noppert
- Social Environment and Health Program, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Constanza X Carney
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Chunyu Liu
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Ashley R M Sapp
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elijah J Watson
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Allison E Aiello
- Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York City, NY, USA
- Department of Epidemiology, Mailman School of Public, Columbia University, New York City, NY, USA
| |
Collapse
|
25
|
Han T, Xu Y, Sun L, Hashimoto M, Wei J. Microglial response to aging and neuroinflammation in the development of neurodegenerative diseases. Neural Regen Res 2024; 19:1241-1248. [PMID: 37905870 PMCID: PMC11467914 DOI: 10.4103/1673-5374.385845] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/30/2023] [Accepted: 07/17/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cellular senescence and chronic inflammation in response to aging are considered to be indicators of brain aging; they have a great impact on the aging process and are the main risk factors for neurodegeneration. Reviewing the microglial response to aging and neuroinflammation in neurodegenerative diseases will help understand the importance of microglia in neurodegenerative diseases. This review describes the origin and function of microglia and focuses on the role of different states of the microglial response to aging and chronic inflammation on the occurrence and development of neurodegenerative diseases, including Alzheimer's disease, Huntington's chorea, and Parkinson's disease. This review also describes the potential benefits of treating neurodegenerative diseases by modulating changes in microglial states. Therefore, inducing a shift from the neurotoxic to neuroprotective microglial state in neurodegenerative diseases induced by aging and chronic inflammation holds promise for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Tingting Han
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yuxiang Xu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lin Sun
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, Henan Province, China
| | - Makoto Hashimoto
- Department of Basic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| |
Collapse
|
26
|
Dong HY, Wang Y, Zhang XY, Zhang M, Yang LH, Zou ZB, Li Y, Xie MM, Yang XW, Wang B. Chemical Constituents from the deep-sea-derived Fungus Aureobasidium melanogenum LUO5. Chem Biodivers 2024; 21:e202400507. [PMID: 38606561 DOI: 10.1002/cbdv.202400507] [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: 03/15/2024] [Indexed: 04/13/2024]
Abstract
Three new C10 and C12 aliphatic δ-lactones (1-3), three new fatty acid methyl esters (4-6), and eight known compounds (7-14) were isolated from the marine Aureobasidium sp. LUO5. Their structures were established by detailed analyses of the NMR, HRESIMS, optical rotation, and ECD data. All isolates were tested for their inhibitory effects on nitric oxide production in LPS-induced BV-2 cells. Notably, compound 4 displayed the strongest inhibitory effect with the IC50 value of 120.3 nM.
Collapse
Affiliation(s)
- Hao-Yu Dong
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Postgraduate Training Base of Guangzhou University of Chinese Medicine, 518104, Shenzhen, China
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Yuan Wang
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Xiao-Yu Zhang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Postgraduate Training Base of Guangzhou University of Chinese Medicine, 518104, Shenzhen, China
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Meng Zhang
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Long-He Yang
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Zheng-Biao Zou
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - You Li
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Ming-Min Xie
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Xian-Wen Yang
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen, 361005, China
| | - Bin Wang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Postgraduate Training Base of Guangzhou University of Chinese Medicine, 518104, Shenzhen, China
| |
Collapse
|
27
|
Fancy NN, Smith AM, Caramello A, Tsartsalis S, Davey K, Muirhead RCJ, McGarry A, Jenkyns MH, Schneegans E, Chau V, Thomas M, Boulger S, Cheung TKD, Adair E, Papageorgopoulou M, Willumsen N, Khozoie C, Gomez-Nicola D, Jackson JS, Matthews PM. Characterisation of premature cell senescence in Alzheimer's disease using single nuclear transcriptomics. Acta Neuropathol 2024; 147:78. [PMID: 38695952 PMCID: PMC11065703 DOI: 10.1007/s00401-024-02727-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/11/2024] [Accepted: 03/28/2024] [Indexed: 05/05/2024]
Abstract
Aging is associated with cell senescence and is the major risk factor for AD. We characterized premature cell senescence in postmortem brains from non-diseased controls (NDC) and donors with Alzheimer's disease (AD) using imaging mass cytometry (IMC) and single nuclear RNA (snRNA) sequencing (> 200,000 nuclei). We found increases in numbers of glia immunostaining for galactosidase beta (> fourfold) and p16INK4A (up to twofold) with AD relative to NDC. Increased glial expression of genes related to senescence was associated with greater β-amyloid load. Prematurely senescent microglia downregulated phagocytic pathways suggesting reduced capacity for β-amyloid clearance. Gene set enrichment and pseudo-time trajectories described extensive DNA double-strand breaks (DSBs), mitochondrial dysfunction and ER stress associated with increased β-amyloid leading to premature senescence in microglia. We replicated these observations with independent AD snRNA-seq datasets. Our results describe a burden of senescent glia with AD that is sufficiently high to contribute to disease progression. These findings support the hypothesis that microglia are a primary target for senolytic treatments in AD.
Collapse
Affiliation(s)
- Nurun N Fancy
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Amy M Smith
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Alessia Caramello
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Stergios Tsartsalis
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Karen Davey
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
- UK Dementia Research Institute Centre, King's College London, London, UK
| | - Robert C J Muirhead
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
- UK Dementia Research Institute Centre, King's College London, London, UK
| | - Aisling McGarry
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marion H Jenkyns
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eleonore Schneegans
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Vicky Chau
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Michael Thomas
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Sam Boulger
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - To Ka Dorcas Cheung
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Emily Adair
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Marianna Papageorgopoulou
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Nanet Willumsen
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Combiz Khozoie
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Johanna S Jackson
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- UK Dementia Research Institute Centre, Imperial College London, London, UK
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- UK Dementia Research Institute Centre, Imperial College London, London, UK.
| |
Collapse
|
28
|
Zhong M, Salberg S, Sampangi S, van der Walt A, Butzkueven H, Mychasiuk R, Jokubaitis V. Leukocyte telomere length in multiple sclerosis: relationship between disability severity and pregnancy history. Mult Scler Relat Disord 2024; 86:105607. [PMID: 38631073 DOI: 10.1016/j.msard.2024.105607] [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: 07/20/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Aging-related processes contribute to neurodegeneration and disability in multiple sclerosis (MS). Biomarkers of biological aging such as leukocyte telomere length (LTL) could help personalise prognosis. Pregnancy has been shown to be protective against disability accumulation in women with MS, though it is unclear if this effect relates to aging mechanisms or LTL. OBJECTIVES This study aimed to cross-sectionally characterise LTL in a cohort of individuals with MS, and to correlate LTL with disability severity and pregnancy history. METHODS We extracted DNA from the whole blood of 501 people with MS in Melbourne, Australia. Expanded Disability Status Scale (EDSS) score and demographic data, as well as pregnancy history for 197 females, were obtained at sample collection. Additional data were extracted from the MSBase Registry. LTL was determined in base pairs (bp) using real-time quantitative polymerase chain reaction. RESULTS A relationship between EDSS score and shorter LTL was robust to multivariable adjustment for demographic and clinical factors including chronological age, with an adjusted LTL reduction per 1.0 increase in EDSS of 97.1 bp (95 % CI = 9.7-184.5 bp, p = 0.030). Adjusted mediation analysis found chronological age accounted for 33.6 % of the relationship between LTL and EDSS score (p = 0.018). In females with pregnancy data, history of pregnancy was associated with older age (median 49.7 vs 33.0 years, p < 0.001). There were no significant relationships between adjusted LTL and any history of pregnancy (LTL increase of 65.3 bp, 95 % CI = -471.0-601.5 bp, p = 0.81) or number of completed pregnancies (LTL increase of 14.6 bp per pregnancy, 95 % CI = -170.3-199.6 bp, p = 0.87). CONCLUSIONS The correlation between LTL and disability independent of chronological age and other factors points to a link between neurological reserve in MS and biological aging, and a potential research target for pathophysiological and therapeutic mechanisms. Although LTL did not significantly differ by pregnancy history, longitudinal analyses could help identify interactions with prospectively captured pregnancy effects.
Collapse
Affiliation(s)
- Michael Zhong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia.
| | - Sabrina Salberg
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Sandeep Sampangi
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia; Department of Neurology, The Alfred Hospital, Melbourne, Australia
| |
Collapse
|
29
|
Tsay HJ, Gan YL, Su YH, Sun YY, Yao HH, Chen HW, Hsu YT, Hsu JTA, Wang HD, Shie FS. Reducing brain Aβ burden ameliorates high-fat diet-induced fatty liver disease in APP/PS1 mice. Biomed Pharmacother 2024; 173:116404. [PMID: 38471275 DOI: 10.1016/j.biopha.2024.116404] [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: 12/06/2023] [Revised: 02/18/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
High-fat diet (HFD)-induced fatty liver disease is a deteriorating risk factor for Alzheimer's disease (AD). Mitigating fatty liver disease has been shown to attenuate AD-like pathology in animal models. However, it remains unclear whether enhancing Aβ clearance through immunotherapy would in turn attenuate HFD-induced fatty liver or whether its efficacy would be compromised by long-term exposure to HFD. Here, the therapeutic potentials of an anti-Aβ antibody, NP106, was investigated in APP/PS1 mice by HFD feeding for 44 weeks. The data demonstrate that NP106 treatment effectively reduced Aβ burden and pro-inflammatory cytokines in HFD-fed APP/PS1 mice and ameliorated HFD-aggravated cognitive impairments during the final 18 weeks of the study. The rejuvenating characteristics of microglia were evident in APP/PS1 mice with NP106 treatment, namely enhanced microglial Aβ phagocytosis and attenuated microglial lipid accumulation, which may explain the benefits of NP106. Surprisingly, NP106 also reduced HFD-induced hyperglycemia, fatty liver, liver fibrosis, and hepatic lipids, concomitant with modifications in the expressions of genes involved in hepatic lipogenesis and fatty acid oxidation. The data further reveal that brain Aβ burden and behavioral deficits were positively correlated with the severity of fatty liver disease and fasting serum glucose levels. In conclusion, our study shows for the first time that anti-Aβ immunotherapy using NP106, which alleviates AD-like disorders in APP/PS1 mice, ameliorates fatty liver disease. Minimizing AD-related pathology and symptoms may reduce the vicious interplay between central AD and peripheral fatty liver disease, thereby highlighting the importance of developing AD therapies from a systemic disease perspective.
Collapse
Affiliation(s)
- Huey-Jen Tsay
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Ling Gan
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Yu-Han Su
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Yu-Yo Sun
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
| | - Heng-Hsiang Yao
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Hui-Wen Chen
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Ying-Ting Hsu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - John Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Horng-Dar Wang
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Feng-Shiun Shie
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC.
| |
Collapse
|
30
|
Wan Y, Lin Y, Tan X, Gong L, Lei F, Wang C, Sun X, Du X, Zhang Z, Jiang J, Liu Z, Wang J, Zhou X, Wang S, Zhou X, Jing P, Zhong Z. Injectable Hydrogel To Deliver Bone Mesenchymal Stem Cells Preloaded with Azithromycin To Promote Spinal Cord Repair. ACS NANO 2024; 18:8934-8951. [PMID: 38483284 DOI: 10.1021/acsnano.3c12402] [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: 03/27/2024]
Abstract
Spinal cord injury is a disease that causes severe damage to the central nervous system. Currently, there is no cure for spinal cord injury. Azithromycin is commonly used as an antibiotic, but it can also exert anti-inflammatory effects by down-regulating M1-type macrophage genes and up-regulating M2-type macrophage genes, which may make it effective for treating spinal cord injury. Bone mesenchymal stem cells possess tissue regenerative capabilities that may help promote the repair of the injured spinal cord. In this study, our objective was to explore the potential of promoting repair in the injured spinal cord by delivering bone mesenchymal stem cells that had internalized nanoparticles preloaded with azithromycin. To achieve this objective, we formulated azithromycin into nanoparticles along with a trans-activating transcriptional activator, which should enhance nanoparticle uptake by bone mesenchymal stem cells. These stem cells were then incorporated into an injectable hydrogel. The therapeutic effects of this formulation were analyzed in vitro using a mouse microglial cell line and a human neuroblastoma cell line, as well as in vivo using a rat model of spinal cord injury. The results showed that the formulation exhibited anti-inflammatory and neuroprotective effects in vitro as well as therapeutic effects in vivo. These results highlight the potential of a hydrogel containing bone mesenchymal stem cells preloaded with azithromycin and trans-activating transcriptional activator to mitigate spinal cord injury and promote tissue repair.
Collapse
Affiliation(s)
- Yujie Wan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
- Ultrasound Medicine Department, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yan Lin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xie Tan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lingyi Gong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fei Lei
- Department of Spine Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Changguang Wang
- DataRevive USA, LLC, 30 W Gude Drive, Rockville, Maryland 20850, United States
| | - Xiaoduan Sun
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingjie Du
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhirong Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Jiang
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhongbing Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingxuan Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoling Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shuzao Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pei Jing
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhirong Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
| |
Collapse
|
31
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. J Neuroinflammation 2024; 21:67. [PMID: 38481312 PMCID: PMC10938757 DOI: 10.1186/s12974-024-03059-x] [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: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles during development as well as in health and disease. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., Box 8057, St. Louis, MO, 63110, USA.
| | - Haowu Jiang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., CB 8054, St. Louis, MO, 63110, USA.
| |
Collapse
|
32
|
Yu PJ, Zhou M, Liu Y, Du J. Senescent T Cells in Age-Related Diseases. Aging Dis 2024; 16:AD.2024.0219. [PMID: 38502582 PMCID: PMC11745454 DOI: 10.14336/ad.2024.0219] [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: 12/08/2023] [Accepted: 02/18/2024] [Indexed: 03/21/2024] Open
Abstract
Age-induced alterations in human immunity are often considered deleterious and are referred to as immunosenescence. The immune system monitors the number of senescent cells in the body, while immunosenescence may represent the initiation of systemic aging. Immune cells, particularly T cells, are the most impacted and involved in age-related immune function deterioration, making older individuals more prone to different age-related diseases. T-cell senescence can impact the effectiveness of immunotherapies that rely on the immune system's function, including vaccines and adoptive T-cell therapies. The research and practice of using senescent T cells as therapeutic targets to intervene in age-related diseases are in their nascent stages. Therefore, in this review, we summarize recent related literature to investigate the characteristics of senescent T cells as well as their formation mechanisms, relationship with various aging-related diseases, and means of intervention. The primary objective of this article is to explore the prospects and possibilities of therapeutically targeting senescent T cells, serving as a valuable resource for the development of immunotherapy and treatment of age-related diseases.
Collapse
Affiliation(s)
- Pei-Jie Yu
- Beijing Anzhen Hospital, Capital Medical University
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education
- Beijing Collaborative Innovative Research Center for Cardiovascular Diseases
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Mei Zhou
- Beijing Anzhen Hospital, Capital Medical University
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education
- Beijing Collaborative Innovative Research Center for Cardiovascular Diseases
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Yan Liu
- Correspondence should be addressed to: Dr. Jie Du () and Dr. Yan Liu (), Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jie Du
- Correspondence should be addressed to: Dr. Jie Du () and Dr. Yan Liu (), Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
33
|
Dennery PA, Yao H. Emerging role of cellular senescence in normal lung development and perinatal lung injury. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:10-16. [PMID: 38567372 PMCID: PMC10987039 DOI: 10.1016/j.pccm.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cellular senescence is a status of irreversible growth arrest, which can be triggered by the p53/p21cip1 and p16INK4/Rb pathways via intrinsic and external factors. Senescent cells are typically enlarged and flattened, and characterized by numerous molecular features. The latter consists of increased surfaceome, increased residual lysosomal activity at pH 6.0 (manifested by increased activity of senescence-associated beta-galactosidase [SA-β-gal]), senescence-associated mitochondrial dysfunction, cytoplasmic chromatin fragment, nuclear lamin b1 exclusion, telomere-associated foci, and the senescence-associated secretory phenotype. These features vary depending on the stressor leading to senescence and the type of senescence. Cellular senescence plays pivotal roles in organismal aging and in the pathogenesis of aging-related diseases. Interestingly, senescence can also both promote and inhibit wound healing processes. We recently report that senescence as a programmed process contributes to normal lung development. Lung senescence is also observed in Down Syndrome, as well as in premature infants with bronchopulmonary dysplasia and in a hyperoxia-induced rodent model of this disease. Furthermore, this senescence results in neonatal lung injury. In this review, we briefly discuss the molecular features of senescence. We then focus on the emerging role of senescence in normal lung development and in the pathogenesis of bronchopulmonary dysplasia as well as putative signaling pathways driving senescence. Finally, we discuss potential therapeutic approaches targeting senescent cells to prevent perinatal lung diseases.
Collapse
Affiliation(s)
- Phyllis A. Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
| |
Collapse
|
34
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. Clin Immunol 2024:109921. [PMID: 38316202 DOI: 10.1016/j.clim.2024.109921] [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: 12/13/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024]
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles in normal brain development, neurodegeneration, and brain cancers. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| | - Haowu Jiang
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine in St Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
35
|
Zhao R. Exercise mimetics: a novel strategy to combat neuroinflammation and Alzheimer's disease. J Neuroinflammation 2024; 21:40. [PMID: 38308368 PMCID: PMC10837901 DOI: 10.1186/s12974-024-03031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
Neuroinflammation is a pathological hallmark of Alzheimer's disease (AD), characterized by the stimulation of resident immune cells of the brain and the penetration of peripheral immune cells. These inflammatory processes facilitate the deposition of amyloid-beta (Aβ) plaques and the abnormal hyperphosphorylation of tau protein. Managing neuroinflammation to restore immune homeostasis and decrease neuronal damage is a therapeutic approach for AD. One way to achieve this is through exercise, which can improve brain function and protect against neuroinflammation, oxidative stress, and synaptic dysfunction in AD models. The neuroprotective impact of exercise is regulated by various molecular factors that can be activated in the same way as exercise by the administration of their mimetics. Recent evidence has proven some exercise mimetics effective in alleviating neuroinflammation and AD, and, additionally, they are a helpful alternative option for patients who are unable to perform regular physical exercise to manage neurodegenerative disorders. This review focuses on the current state of knowledge on exercise mimetics, including their efficacy, regulatory mechanisms, progress, challenges, limitations, and future guidance for their application in AD therapy.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, Yangzhou, China.
| |
Collapse
|
36
|
Shi M, Chu F, Zhu F, Zhu J. Peripheral blood amyloid-β involved in the pathogenesis of Alzheimer's disease via impacting on peripheral innate immune cells. J Neuroinflammation 2024; 21:5. [PMID: 38178136 PMCID: PMC10765910 DOI: 10.1186/s12974-023-03003-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
A key pathological factor of Alzheimer's disease (AD), the most prevalent form of age-related dementia in the world, is excessive β-amyloid protein (Aβ) in extracellular aggregation in the brain. And in the peripheral blood, a large amount of Aβ is derived from platelets. So far, the causality between the levels of peripheral blood Aβ and its aggregation in the brain, particularly the role of the peripheral blood Aβ in the pathology of AD, is still unclear. And the relation between the peripheral blood Aβ and tau tangles of brain, another crucial pathologic factor contributing to the pathogenesis of AD, is also ambiguous. More recently, the anti-Aβ monoclonal antibodies are approved for treatment of AD patients through declining the peripheral blood Aβ mechanism of action to enhance plasma and central nervous system (CNS) Aβ clearance, leading to a decrease Aβ burden in brain and improving cognitive function, which clearly indicates that the levels of the peripheral blood Aβ impacted on the Aβ burden in brain and involved in the pathogenesis of AD. In addition, the role of peripheral innate immune cells in AD remains mostly unknown and the results obtained were controversial. In the present review, we summarize recent studies on the roles of peripheral blood Aβ and the peripheral innate immune cells in the pathogenesis of AD. Finally, based on the published data and our own work, we believe that peripheral blood Aβ plays an important role in the development and progression of AD by impacting on the peripheral innate immune cells.
Collapse
Affiliation(s)
- Mingchao Shi
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Fengna Chu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Feiqi Zhu
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden.
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China.
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.
- Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden.
| |
Collapse
|
37
|
Jimenez-Gómez N, López-Suárez A, Haro S, Fernández-González P, Monserrat J, Eraña-Tomás I, Cuevas-Santos J, Rodríguez-Luna A, Ortega MA, Gómez-Sánchez MJ, Díaz D, Jaén-Olasolo P, Álvarez-Mon M. Immunomodulation with AM3 and antioxidants creates an adequate framework for skin repair and decreases the monocyte proinflammatory stage in smoker women. Biomed Pharmacother 2024; 170:115929. [PMID: 38070248 DOI: 10.1016/j.biopha.2023.115929] [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: 07/27/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 01/10/2024] Open
Abstract
Smoking has been considering a crucial factor in promoting skin and systemic aging that is associated with the development of a low-level, systemic, chronic inflammation known as "inflammaging" in which monocytes play a pivotal role. Our aim was to investigate the effects of AM3 plus antioxidants vs placebo in the activation status, function of monocytes and cutaneous aging parameters in healthy smoker middle-aged women. A total of 32 women were 1:1 randomly assigned to AM3 plus antioxidants or placebo for three months. Peripheral mononuclear blood cells and cutaneous biopsy were obtained and flow cytometry and immunohistological studies, respectively, were performed before and after the treatment. AM3 plus antioxidants treatment compared with placebo significantly reduced the monocyte production of the proinflammatory interleukin 1 (IL-1), tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) cytokines as well as increased the regulatory IL-10 in middle-aged smoker women. Furthermore, AM3 and antioxidants did not modify ROS production by monocytes and granulocytes but increased their phagocytic activity. The active combination also stimulated a significative increase in reticular dermis depth as well as an increase in the expression of CD117 and CD31. Thus, AM3 and antioxidants treatment reduces the systemic proinflammatory monocyte disturbance of heathy smoker middle-aged women and encourage skin repair mechanisms.
Collapse
Affiliation(s)
| | - Andrea López-Suárez
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital "Príncipe de Asturias", Alcalá de Henares, 28805 Madrid, Spain
| | - Sergio Haro
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid 28801, Spain
| | - Pablo Fernández-González
- Dermatology Department, Ramón y Cajal Hospital, 28034 Madrid, Spain; Medical department, Cantabria Labs, 28043 Madrid, Spain.
| | - Jorge Monserrat
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital "Príncipe de Asturias", Alcalá de Henares, 28805 Madrid, Spain
| | - Itziar Eraña-Tomás
- Pathology Department, Fundación Jimenez Díaz Hospital, 28040 Madrid, Spain
| | - Jesus Cuevas-Santos
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid 28801, Spain
| | - Azahara Rodríguez-Luna
- Faculty of Health Sciences, Universidad Loyola Andalucía, Seville, Spain; Department of Pharmacology, Faculty of Pharmacy, University of Seville, Profesor García González Street Seville, 41012, Spain
| | - Miguel A Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid 28801, Spain; Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | | | - David Díaz
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital "Príncipe de Asturias", Alcalá de Henares, 28805 Madrid, Spain
| | - Pedro Jaén-Olasolo
- Dermatology Department, Ramón y Cajal Hospital, 28034 Madrid, Spain; Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid 28801, Spain
| | - Melchor Álvarez-Mon
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital "Príncipe de Asturias", Alcalá de Henares, 28805 Madrid, Spain; Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid 28801, Spain
| |
Collapse
|
38
|
Wang G, Wang L, Wang X, Ye H, Ni W, Shao W, Dai C, Liu B. Low-intensity exercise training increases systolic function of heart and MHCII low cardiac resident macrophages. Heliyon 2023; 9:e22915. [PMID: 38076084 PMCID: PMC10703626 DOI: 10.1016/j.heliyon.2023.e22915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 08/11/2024] Open
Abstract
Physical activities have beneficial effects on cardiovascular health, although the specific mechanisms are largely unknown. Cardiac resident macrophages (cMacs) and the distribution of their subsets are critical regulators for maintaining cardiovascular health and cardiac functions in both steady and inflammatory states. Therefore, we investigated the subsets of cMacs in mice after low-intensity exercise training to elucidate the exercise-induced dynamic changes of cMacs and the benefits of exercise for the heart. The mice were subjected to treadmill running exercise five days per week for five weeks using a low-intensity exercise training protocol. Low-intensity exercise training resulted in a suppression of body weight gain in mice and a significant increase in the ejection fraction, a parameter that represents the systolic function of the heart. Low-intensity exercise training induced the alterations in the transcriptome of the heart, which are associated with muscle contraction and mitochondrial function. Furthermore, low-intensity exercise training did not alter the number of lymphocyte antigen 6 complex, locus C1 (Ly6c)- cMacs but instead remodeled the distributions of Ly6c- cMac subsets. We observed an increase in the percentage of major histocompatibility complex class II (MHCII)low cMacs and a decrease in the percentage of MHCIIhigh cMacs in the heart after low-intensity exercise training. Therefore, the benefits of exercise for cardiovascular fitness might be associated with the redistribution of cMac subsets and the enhancement of the ejection fraction.
Collapse
Affiliation(s)
| | | | - Xuchao Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Heng Ye
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Wei Ni
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Wei Shao
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Cuilian Dai
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| | - Binbin Liu
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Jinshan Road 2999, Xiamen, 361015, China
| |
Collapse
|
39
|
Zhang Y, Xie CL, Wang Y, He XW, Xie MM, Li Y, Zhang K, Zou ZB, Yang LH, Xu R, Yang XW. Penidihydrocitrinins A-C: New Polyketides from the Deep-Sea-Derived Penicillium citrinum W17 and Their Anti-Inflammatory and Anti-Osteoporotic Bioactivities. Mar Drugs 2023; 21:538. [PMID: 37888473 PMCID: PMC10608093 DOI: 10.3390/md21100538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Three new polyketides (penidihydrocitrinins A-C, 1-3) and fourteen known compounds (4-17) were isolated from the deep-sea-derived Penicillium citrinum W17. Their structures were elucidated by comprehensive analyses of 1D and 2D NMR, HRESIMS, and ECD calculations. Compounds 1-17 were evaluated for their anti-inflammatory and anti-osteoporotic bioactivities. All isolates exhibited significant inhibitory effects on LPS-stimulated nitric oxide production in murine brain microglial BV-2 cells in a dose-response manner. Notably, compound 14 displayed the strongest effect with the IC50 value of 4.7 µM. Additionally, compounds 6, 7, and 8 significantly enhanced osteoblast mineralization, which was comparable to that of the positive control, purmorphamine. Furthermore, these three compounds also suppressed osteoclastogenesis in a dose-dependent manner under the concentrations of 2.5 μM, 5.0 μM, and 10 μM.
Collapse
Affiliation(s)
- Yong Zhang
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - Chun-Lan Xie
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
- School of Medicine, Xiamen University, South Xiangan Road, Xiamen 361005, China
| | - Yuan Wang
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - Xi-Wen He
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - Ming-Min Xie
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - You Li
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - Kai Zhang
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - Zheng-Biao Zou
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - Long-He Yang
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| | - Ren Xu
- School of Medicine, Xiamen University, South Xiangan Road, Xiamen 361005, China
| | - Xian-Wen Yang
- Key Laboratory of Marine Genetic Resources, Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China; (Y.Z.); (C.-L.X.); (Y.W.); (X.-W.H.); (M.-M.X.); (Y.L.); (K.Z.); (Z.-B.Z.)
| |
Collapse
|
40
|
Lopes JR, Zhang X, Mayrink J, Tatematsu BK, Guo L, LeServe DS, Abou-El-Hassan H, Rong F, Dalton MJ, Oliveira MG, Lanser TB, Liu L, Butovsky O, Rezende RM, Weiner HL. Nasal administration of anti-CD3 monoclonal antibody ameliorates disease in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2023; 120:e2309221120. [PMID: 37669383 PMCID: PMC10500187 DOI: 10.1073/pnas.2309221120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Emerging evidence suggests that dysregulation of neuroinflammation, particularly that orchestrated by microglia, plays a significant role in the pathogenesis of Alzheimer's disease (AD). Danger signals including dead neurons, dystrophic axons, phosphorylated tau, and amyloid plaques alter the functional phenotype of microglia from a homeostatic (M0) to a neurodegenerative or disease-associated phenotype, which in turn drives neuroinflammation and promotes disease. Thus, therapies that target microglia activation constitute a unique approach for treating AD. Here, we report that nasally administered anti-CD3 monoclonal antibody in the 3xTg AD mouse model reduced microglial activation and improved cognition independent of amyloid beta deposition. In addition, gene expression analysis demonstrated decreased oxidative stress, increased axogenesis and synaptic organization, and metabolic changes in the hippocampus and cortex of nasal anti-CD3 treated animals. The beneficial effect of nasal anti-CD3 was associated with the accumulation of T cells in the brain where they were in close contact with microglial cells. Taken together, our findings identify nasal anti-CD3 as a unique form of immunotherapy to treat Alzheimer's disease independent of amyloid beta targeting.
Collapse
Affiliation(s)
- Juliana R. Lopes
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Xiaoming Zhang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Julia Mayrink
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Bruna K. Tatematsu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Lydia Guo
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Danielle S. LeServe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Hadi Abou-El-Hassan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Felipe Rong
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Maria J. Dalton
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Marilia G. Oliveira
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Toby B. Lanser
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| |
Collapse
|
41
|
Li SJ, Shi JJ, Mao CY, Zhang C, Xu YF, Fan Y, Hu ZW, Yu WK, Hao XY, Li MJ, Li JD, Ma DR, Guo MN, Zuo CY, Liang YY, Xu YM, Wu J, Sun SL, Wang YG, Shi CH. Identifying causal genes for migraine by integrating the proteome and transcriptome. J Headache Pain 2023; 24:111. [PMID: 37592229 PMCID: PMC10433568 DOI: 10.1186/s10194-023-01649-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND While previous genome-wide association studies (GWAS) have identified multiple risk variants for migraine, there is a lack of evidence about how these variants contribute to the development of migraine. We employed an integrative pipeline to efficiently transform genetic associations to identify causal genes for migraine. METHODS We conducted a proteome-wide association study (PWAS) by combining data from the migraine GWAS data with proteomic data from the human brain and plasma to identify proteins that may play a role in the risk of developing migraine. We also combined data from GWAS of migraine with a novel joint-tissue imputation (JTI) prediction model of 17 migraine-related human tissues to conduct transcriptome-wide association studies (TWAS) together with the fine mapping method FOCUS to identify disease-associated genes. RESULTS We identified 13 genes in the human brain and plasma proteome that modulate migraine risk by regulating protein abundance. In addition, 62 associated genes not reported in previous migraine TWAS studies were identified by our analysis of migraine using TWAS and fine mapping. Five genes including ICA1L, TREX1, STAT6, UFL1, and B3GNT8 showed significant associations with migraine at both the proteome and transcriptome, these genes are mainly expressed in ependymal cells, neurons, and glial cells, and are potential target genes for prevention of neuronal signaling and inflammatory responses in the pathogenesis of migraine. CONCLUSIONS Our proteomic and transcriptome findings have identified disease-associated genes that may give new insights into the pathogenesis and potential therapeutic targets for migraine.
Collapse
Affiliation(s)
- Shuang-Jie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jing-Jing Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Cheng-Yuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chan Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Ya-Fang Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yu Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zheng-Wei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Wen-Kai Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiao-Yan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Meng-Jie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jia-di Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dong-Rui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Meng-Nan Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chun-Yan Zuo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yuan-Yuan Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shi-Lei Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yong-Gang Wang
- Headache Center, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| | - Chang-He Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
42
|
Mohapatra L, Mishra D, Shiomurti Tripathi A, Kumar Parida S. Immunosenescence as a convergence pathway in neurodegeneration. Int Immunopharmacol 2023; 121:110521. [PMID: 37385122 DOI: 10.1016/j.intimp.2023.110521] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
Immunity refers to the body's defense mechanism to protect itself against illness or to produce antibodies against pathogens. Senescence is a cellular phenomenon that integrates a sustainable growth restriction, other phenotypic abnormalities and including a pro-inflammatory secretome. It is highly involved in regulating developmental stages, tissue homeostasis, and tumor proliferation monitoring. Contemporary experimental reports imply that abolition of senescent cells employing evolved genetic and therapeutic approaches augment the chances of survival and boosts the health span of an individual. Immunosenescence is considered as a process in which dysfunction of the immune system occurs with aging and greatly includes remodeling of lymphoid organs. This in turn causes fluctuations in the immune function of the elderly that has strict relation with the expansion of autoimmune diseases, infections, malignant tumors and neurodegenerative disorders. The interaction of the nervous and immune systems during aging is marked by bi-directional influence and mutual correlation of variations. The enhanced systemic inflammatory condition in the elderly, and the neuronal immune cell activity can be modulated by inflamm-aging and peripheral immunosenescence resulting in chronic low-grade inflammatory processes in the central Nervous system known as neuro-inflammaging. For example, glia excitation by cytokines and glia pro-inflammatory productions contribute significantly to memory injury as well as in acute systemic inflammation, which is associated with high levels of Tumor necrosis factor -α and a rise in cognitive decline. In recent years its role in the pathology of Alzheimer's disease has caught research interest to a large extent. This article reviews the connection concerning the immune and nervous systems and highlights how immunosenescence and inflamm-aging can affect neurodegenerative disorders.
Collapse
Affiliation(s)
- Lucy Mohapatra
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh Sector-125, Noida, 201313, India.
| | - Deepak Mishra
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh Sector-125, Noida, 201313, India
| | | | | |
Collapse
|
43
|
Yang G, Tong Y, Wang X, Zhao C, Ba Z, Ahelijiang R, Liu X, Gao W, Zhao Y, Gu Y, Yang J, Xu Y. Guizhi Fuling capsule relieves memory deficits by inhibition of microglial neuroinflammation through blocking JAK2/STAT3 pathway in presenilin1/2 conditional double knockout mice. Front Immunol 2023; 14:1185570. [PMID: 37465679 PMCID: PMC10350565 DOI: 10.3389/fimmu.2023.1185570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
Chronic neuroinflammation has been regarded as an important part of the pathological initiation of Alzheimer's disease (AD), which is associated with the regulation of microglial activation. Preventing microglial activation to inhibit neuroinflammation may become a potential target for the treatment of neurodegenerative diseases. Guizhi Fuling capsule (GZFL) has a strong repression on inflammatory responses. Here, the presenilin1/2 conditional double knockout (PS cDKO) mice, a well-established mouse model of AD, were divided into: WT mice (WT), WT mice+GZFL (WT+GZFL), PS cDKO mice (cDKO), and PS cDKO mice+GZFL (cDKO+GZFL). Mice in the WT+GZFL and cDKO+GZFL group were fed standard chow containing 2000 ppm GZFL for 90 days. After 60 days of GZFL treatment, mice were given to behavioral tests for 30 days in order to explore the effects of GZFL on cognitive and motor function. Then, mice were sacrificed for examining the effects of GZFL on inflammation. Furthermore, primary microglia were obtained from neonatal Sprague-Dawley rats and pretreated with or without GZFL (50 μg/ml) for 1 h in the absence or presence of lipopolysaccharide (LPS) (100 ng/ml) stimulation to speculate whether the underlying mechanism of GZFL's anti-inflammatory potential was closely associated with Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Our findings indicated that GZFL has the ability to alleviate memory deficits in PS cDKO mice, which attributes to the improvement of neuroinflammation by inhibiting microglial activation and the levels of pro-inflammatory mediators. In addition, GZFL could inverse the tau hyperphosphorylation and the lessened expression of synaptic proteins in hippocampus of PS cDKO mice. Furthermore, GZFL prevented LPS-induced neuroinflammatory responses in primary microglia by decreasing the levels of pro-inflammatory mediators. It is noteworthy that therapeutic effects of GZFL on memory impairment are depended on the inhibition of neuroinflammatory responses by the blockage of JAK2/STAT3 signaling pathway. Taken together, GZFL may be an effective compound Chinese medicine for the improvement and postponement of neurodegenerative progression in AD.
Collapse
Affiliation(s)
- Guang Yang
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuting Tong
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xingyu Wang
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyi Zhao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongtao Ba
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Reaila Ahelijiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinjuan Liu
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Waimao Gao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhao
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yining Gu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Yangzhi Rehabilitation Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianmei Yang
- Department of Traditional Chinese Medicine, Shanghai Xuhui District Central Hospital, Shanghai, China
| | - Ying Xu
- Department of Physiology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
de la Fuente AG, Pelucchi S, Mertens J, Di Luca M, Mauceri D, Marcello E. Novel therapeutic approaches to target neurodegeneration. Br J Pharmacol 2023; 180:1651-1673. [PMID: 36965025 PMCID: PMC10952850 DOI: 10.1111/bph.16078] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023] Open
Abstract
Ageing is the main risk factor common to most primary neurodegenerative disorders. Indeed, age-related brain alterations have been long considered to predispose to neurodegeneration. Although protein misfolding and the accumulation of toxic protein aggregates have been considered as causative events in neurodegeneration, several other biological pathways affected by brain ageing also contribute to pathogenesis. Here, we discuss the evidence showing the involvement of the mechanisms controlling neuronal structure, gene expression, autophagy, cell metabolism and neuroinflammation in the onset and progression of neurodegenerative disorders. Furthermore, we review the therapeutic strategies currently under development or as future approaches designed to normalize these pathways, which may then increase brain resilience to cope with toxic protein species. In addition to therapies targeting the insoluble protein aggregates specifically associated with each neurodegenerative disorder, these novel pharmacological approaches may be part of combined therapies designed to rescue brain function.
Collapse
Affiliation(s)
- Alerie G. de la Fuente
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
- Instituto de Neurociencias CSIC‐UMHAlicanteSpain
- Wellcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
| | - Jerome Mertens
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Daniela Mauceri
- Institute of Anatomy and Cell BiologyDepartment of Molecular and Cellular Neuroscience, University of MarburgMarburgGermany
- Department of NeurobiologyInterdisciplinary Centre for Neurosciences (IZN), Heidelberg UniversityHeidelbergGermany
| | - Elena Marcello
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| |
Collapse
|
45
|
Pilipović I, Stojić-Vukanić Z, Prijić I, Jasnić N, Djordjević J, Leposavić G. β-Adrenoceptor Blockade Moderates Neuroinflammation in Male and Female EAE Rats and Abrogates Sexual Dimorphisms in the Major Neuroinflammatory Pathways by Being More Efficient in Males. Cell Mol Neurobiol 2023; 43:1237-1265. [PMID: 35798933 PMCID: PMC11414456 DOI: 10.1007/s10571-022-01246-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/18/2022] [Indexed: 11/03/2022]
Abstract
Our previous studies showed more severe experimental autoimmune encephalomyelitis (EAE) in male compared with female adult rats, and moderating effect of propranolol-induced β-adrenoceptor blockade on EAE in females, the effect associated with transcriptional stimulation of Nrf2/HO-1 axis in spinal cord microglia. This study examined putative sexual dimorphism in propranolol action on EAE severity. Propranolol treatment beginning from the onset of clinical EAE mitigated EAE severity in rats of both sexes, but to a greater extent in males exhibiting higher noradrenaline levels and myeloid cell β2-adrenoceptor expression in spinal cord. This correlated with more prominent stimulatory effects of propranolol not only on CX3CL1/CX3CR1/Nrf2/HO-1 cascade, but also on Stat3/Socs3 signaling axis in spinal cord microglia/myeloid cells (mirrored in the decreased Stat3 and the increased Socs3 expression) from male rats compared with their female counterparts. Propranolol diminished the frequency of activated cells among microglia, increased their phagocyting/endocyting capacity, and shifted cytokine secretory profile of microglia/blood-borne myeloid cells towards an anti-inflammatory/neuroprotective phenotype. Additionally, it downregulated the expression of chemokines (CCL2, CCL19/21) driving T-cell/monocyte trafficking into spinal cord. Consequently, in propranolol-treated rats fewer activated CD4+ T cells and IL-17+ T cells, including CD4+IL17+ cells coexpressing IFN-γ/GM-CSF, were recovered from spinal cord of propranolol-treated rats compared with sex-matched saline-injected controls. All the effects of propranolol were more prominent in males. The study as a whole disclosed that sexual dimorphism in multiple molecular mechanisms implicated in EAE development may be responsible for greater severity of EAE in male rats and sexually dimorphic action of substances affecting them. Propranolol moderated EAE severity more effectively in male rats, exhibiting greater spinal cord noradrenaline (NA) levels and myeloid cell β2-adrenoceptor (β2-AR) expression than females. Propranolol affected CX3CR1/Nrf2/HO-1 and Stat3/Socs3 signaling axes in myeloid cells, favored their anti-inflammatory/neuroprotective phenotype and, consequently, reduced Th cell reactivation and differentiation into highly pathogenic IL-17/IFN-γ/GM-CSF-producing cells.
Collapse
Affiliation(s)
- Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Ivana Prijić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Nebojša Jasnić
- Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Jelena Djordjević
- Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221, Belgrade, Serbia.
| |
Collapse
|
46
|
Thornton T, Mills D, Bliss E. Capsaicin: A Potential Treatment to Improve Cerebrovascular Function and Cognition in Obesity and Ageing. Nutrients 2023; 15:nu15061537. [PMID: 36986266 PMCID: PMC10057869 DOI: 10.3390/nu15061537] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Impaired cognition is the primary symptom of dementia, which can lead to functional disability and reduced quality of life among an increasingly ageing population. Ageing is associated with increased oxidative stress, chronic low-grade systemic inflammation, and endothelial dysfunction, which reduces cerebrovascular function leading to cognitive decline. Chronic low-grade systemic inflammatory conditions, such as obesity, exacerbate this decline beyond normal ageing and predispose individuals to neurodegenerative diseases, such as dementia. Capsaicin, the major pungent molecule of chilli, has recently demonstrated improvements in cognition in animal models via activation of the transient receptor potential vanilloid channel 1 (TRPV1). Capsaicin-induced TRPV1 activation reduces adiposity, chronic low-grade systemic inflammation, and oxidative stress, as well as improves endothelial function, all of which are associated with cerebrovascular function and cognition. This review examines the current literature on capsaicin and Capsimax, a capsaicin supplement associated with reduced gastrointestinal irritation compared to capsaicin. Acute and chronic capsaicin treatment can improve cognition in animals. However, studies adequately assessing the effects of capsaicin on cerebrovascular function, and cognition in humans do not exist. Capsimax may be a potentially safe therapeutic intervention for future clinical trials testing the effects of capsaicin on cerebrovascular function and cognition.
Collapse
Affiliation(s)
- Tammy Thornton
- School of Health and Medical Sciences, University of Southern Queensland, Ipswich, QLD 4305, Australia
| | - Dean Mills
- School of Health and Medical Sciences, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Respiratory and Exercise Physiology Research Group, School of Health and Medical Sciences, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Centre for Health Research, Institute for Resilient Regions, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Molecular Biomarkers Research Group, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Edward Bliss
- School of Health and Medical Sciences, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Respiratory and Exercise Physiology Research Group, School of Health and Medical Sciences, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Centre for Health Research, Institute for Resilient Regions, University of Southern Queensland, Ipswich, QLD 4305, Australia
- Molecular Biomarkers Research Group, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| |
Collapse
|
47
|
Kashif M, Waseem M, Vijendra PD, Pandurangan AK. Protective Effects of Cannabis in Neuroinflammation-Mediated Alzheimer's Disease. ADVANCES IN MEDICAL DIAGNOSIS, TREATMENT, AND CARE 2023:48-75. [DOI: 10.4018/978-1-6684-5652-1.ch002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
In recent years, Alzheimer's disease (AD) has been recognized as an age-related neurological disorder wherein neurons degenerate and exhibit abnormal structure and function. Aging is the primary factor in the progression of AD from mild to severe cognitive impairment. No effective targeted therapies are presently available, and treatment is limited to symptomatic management. The neuropathologic hallmarks of the disease include the accumulation of amyloid-beta (Aβ) plaques in brain tissues and the aggregation of hyperphosphorylated-tau proteins (tangles) within neurons. Associated hyperactivation of neuroinflammation results in release of inflammatory molecules from neurons, microglia, and astrocytes, which have been linked with neuronal loss and the worsening neurodegeneration. The anti-inflammatory and neuroprotective properties of cannabis-based medicines may offer benefits in delaying the progression of neurodegenerative diseases including AD. This chapter explores the role of cannabinoids in countering neuroinflammation-mediated AD pathology.
Collapse
Affiliation(s)
- Mohd Kashif
- B.S. Abdur Rahman Crescent Institute of Science and Technology, India
| | | | | | | |
Collapse
|
48
|
He CY, Tian DY, Chen SH, Jin WS, Cheng Y, Xin JY, Li WW, Zeng GH, Tan CR, Jian JM, Fan DY, Ren JR, Liu YH, Wang YJ, Zeng F. Elevated Levels of Naturally-Occurring Autoantibodies Against the Extracellular Domain of p75NTR Aggravate the Pathology of Alzheimer's Disease. Neurosci Bull 2023; 39:261-272. [PMID: 35974288 PMCID: PMC9905456 DOI: 10.1007/s12264-022-00936-4] [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: 03/01/2022] [Accepted: 05/24/2022] [Indexed: 10/15/2022] Open
Abstract
The extracellular domain (p75ECD) of p75 neurotrophin receptor (p75NTR) antagonizes Aβ neurotoxicity and promotes Aβ clearance in Alzheimer's disease (AD). The impaired shedding of p75ECD is a key pathological process in AD, but its regulatory mechanism is largely unknown. This study was designed to investigate the presence and alterations of naturally-occurring autoantibodies against p75ECD (p75ECD-NAbs) in AD patients and their effects on AD pathology. We found that the cerebrospinal fluid (CSF) level of p75ECD-NAbs was increased in AD, and negatively associated with the CSF levels of p75ECD. Transgenic AD mice actively immunized with p75ECD showed a lower level of p75ECD and more severe AD pathology in the brain, as well as worse cognitive functions than the control groups, which were immunized with Re-p75ECD (the reverse sequence of p75ECD) and phosphate-buffered saline, respectively. These findings demonstrate the impact of p75ECD-NAbs on p75NTR/p75ECD imbalance, providing a novel insight into the role of autoimmunity and p75NTR in AD.
Collapse
Affiliation(s)
- Chen-Yang He
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, 610000, China
| | - Ding-Yuan Tian
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Si-Han Chen
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Department of Neurology, Nanchong Central Hospital, The Second Clinical Medical School, North Sichuan Medical College, Nanchong, 637000, China
| | - Wang-Sheng Jin
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, 400042, China
| | - Yuan Cheng
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Jia-Yan Xin
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China
| | - Wei-Wei Li
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, 610000, China
| | - Gui-Hua Zeng
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, 400042, China
| | - Cheng-Rong Tan
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, 400042, China
| | - Jie-Ming Jian
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Dong-Yu Fan
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, 857000, China
| | - Jun-Rong Ren
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yu-Hui Liu
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, 400042, China
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- The Institute of Brain and Intelligence, Third Military Medical University, Chongqing, 400042, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Fan Zeng
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, 400042, China.
| |
Collapse
|
49
|
Bao H, Shen Y. Unmasking BACE1 in aging and age-related diseases. Trends Mol Med 2023; 29:99-111. [PMID: 36509631 DOI: 10.1016/j.molmed.2022.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022]
Abstract
The beta-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) has long been considered a conventional target for Alzheimer's disease (AD). Unfortunately, AD clinical trials of most BACE1 inhibitors were discontinued due to ineffective cognitive improvement or safety challenges. Recent studies investigating the involvement of BACE1 in metabolic, vascular, and immune functions have indicated a role in aging, diabetes, hypertension, and cancer. These novel BACE1 functions have helped to identify new 'druggable' targets for BACE1 against aging comorbidities. In this review, we discuss BACE1 regulation during aging, and then provide recent insights into its enzymatic and nonenzymatic involvement in aging and age-related diseases. Our study not only proposes the perspective of BACE1's actions in various systems, but also provides new directions for using BACE1 inhibitors and modulators to delay aging and to treat age-related diseases.
Collapse
Affiliation(s)
- Hong Bao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Provincial Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 230026, China; CAS Key Laboratory of Brain Function and Disease, Division of Biological and Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
50
|
Yao H, Wallace J, Peterson AL, Scaffa A, Rizal S, Hegarty K, Maeda H, Chang JL, Oulhen N, Kreiling JA, Huntington KE, De Paepe ME, Barbosa G, Dennery PA. Timing and cell specificity of senescence drives postnatal lung development and injury. Nat Commun 2023; 14:273. [PMID: 36650158 PMCID: PMC9845377 DOI: 10.1038/s41467-023-35985-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Senescence causes age-related diseases and stress-related injury. Paradoxically, it is also essential for organismal development. Whether senescence contributes to lung development or injury in early life remains unclear. Here, we show that lung senescence occurred at birth and decreased throughout the saccular stage in mice. Reducing senescent cells at this stage disrupted lung development. In mice (<12 h old) exposed to hyperoxia during the saccular stage followed by air recovery until adulthood, lung senescence increased particularly in type II cells and secondary crest myofibroblasts. This peaked during the alveolar stage and was mediated by the p53/p21 pathway. Decreasing senescent cells during the alveolar stage attenuated hyperoxia-induced alveolar and vascular simplification. Conclusively, early programmed senescence orchestrates postnatal lung development whereas later hyperoxia-induced senescence causes lung injury through different mechanisms. This defines the ontogeny of lung senescence and provides an optimal therapeutic window for mitigating neonatal hyperoxic lung injury by inhibiting senescence.
Collapse
Affiliation(s)
- Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA.
| | - Joselynn Wallace
- Center for Computational Biology of Human Disease and Center for Computation and Visualization, Brown University, Providence, RI, 02912, USA
| | - Abigail L Peterson
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Alejandro Scaffa
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Salu Rizal
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Katy Hegarty
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Hajime Maeda
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Jason L Chang
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Nathalie Oulhen
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Jill A Kreiling
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Kelsey E Huntington
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Monique E De Paepe
- Department of Pathology, Women and Infants Hospital, Providence, RI, 02905, USA
| | - Guilherme Barbosa
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Phyllis A Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA.
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.
| |
Collapse
|