1
|
Lin FV, Simmons JM, Turnbull A, Zuo Y, Conwell Y, Wang KH. Cross-Species Framework for Emotional Well-Being and Brain Aging: Lessons From Behavioral Neuroscience. JAMA Psychiatry 2025:2833240. [PMID: 40332879 DOI: 10.1001/jamapsychiatry.2025.0581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Importance Emotional well-being (EWB) is an emerging therapeutic target for managing and preventing symptoms associated with Alzheimer disease and related dementias (ADRD). However, more research is needed to establish causal inferences between brain changes, EWB, and behavioral changes observed in typical aging and ADRD. Observations This article presents a framework for using a cross-species behavioral neuroscience approach to study EWB and brain aging, adopting a well-established biobehavioral model that highlights the reciprocal roles of brain changes, EWB, and ADRD symptoms. First, the challenges and opportunities in this field are reviewed. Then, a practical solution to improve comparability between animal and human studies is proposed. Conclusions and Relevance The goal is to draw comprehensive parallels and distinctions that could enhance the understanding of the mechanisms linking brain aging, EWB, and ADRD symptomatic disturbances across different species.
Collapse
Affiliation(s)
- F Vankee Lin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Janine M Simmons
- Office of Behavioral and Social Sciences Research, National Institutes of Health, Bethesda, Maryland
| | - Adam Turnbull
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Yi Zuo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
| | - Yeates Conwell
- Department of Psychiatry, University of Rochester, Rochester, New York
| | - Kuan Hong Wang
- Department of Neuroscience, University of Rochester, Rochester, New York
| |
Collapse
|
2
|
Crispens C, Fleckenstein E, Wilken-Schmitz A, Weber S, Gröger M, Hoffmann A, Radermacher P, Reiss LK, Talbot SR, Kästner L, Köhler K, Zacharowski K, von Knethen A, Heinicke U. Sex- and age-related differences in LPS-induced lung injury: establishing a mouse intensive care unit. Intensive Care Med Exp 2025; 13:48. [PMID: 40327214 PMCID: PMC12055714 DOI: 10.1186/s40635-025-00756-6] [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: 02/05/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Mouse models are widely used to establish new therapy concepts for acute lung injury, but the transfer of therapeutic approaches into the intensive care unit often failed. To establish a mouse intensive care unit to adequately reflect the patient's situation and to investigate sex- and age-related differences in response to lipopolysaccharide. METHODS For the establishment of a mouse intensive care unit, young (2-3 months) and old (15-18 months) mice of both sexes received continuous respiratory and cardiovascular monitoring for 6 h. Mimicking an acute lung injury by intratracheal lipopolysaccharide stimulation for 6 or 24 h, the impact of sex and age on survival and physiological parameters was evaluated. RESULTS The establishment revealed sex- and age-related differences in physiological responses during mechanical ventilation, with old males requiring more noradrenaline to maintain stable hemodynamics. While young mice, irrespective of sex, developed acute lung injury 24 h after lipopolysaccharide administration, old mice exhibited a rapid systemic response, showing signs of lactic acidosis and endotoxemia. Among these, old females had the highest mortality risk, whereas in old males, mechanical ventilation provided effective support, contributing to improved survival outcomes. CONCLUSIONS We successfully established a mouse intensive care unit that integrated all critical aspects of a human intensive care unit simultaneously. By highlighting sex- and age-related differences following lipopolysaccharide stimulation and mechanical ventilation, our study underscored the need for diversity in preclinical models to improve translation of findings on critical illnesses like acute lung injury into clinical settings.
Collapse
Affiliation(s)
- Chantal Crispens
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Emilia Fleckenstein
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Annett Wilken-Schmitz
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Sandra Weber
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, Helmholtzstrasse 8-1, Ulm, Germany
| | - Michael Gröger
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, Helmholtzstrasse 8-1, Ulm, Germany
| | - Andrea Hoffmann
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, Helmholtzstrasse 8-1, Ulm, Germany
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University, Helmholtzstrasse 8-1, Ulm, Germany
| | - Lucy Kathleen Reiss
- Department of Pharmacology and Toxicology, RWTH Aachen University, 52074, Aachen, Germany
| | - Steven R Talbot
- Hannover Medical School, Institute for Laboratory Animal Science, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Laura Kästner
- Institute of Veterinary Pathology, Justus Liebig University, Frankfurter Str. 96, 35392, Giessen, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University, Frankfurter Str. 96, 35392, Giessen, Germany
| | - Kai Zacharowski
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Andreas von Knethen
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Ulrike Heinicke
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
3
|
Gao V, Chlebowicz J, Gaskin K, Briano JA, Komer LE, Pineda A, Jhalani S, Ahmad S, Uwaifo E, Black LS, Haller JE, Przedborski S, Lane DA, Zhang S, Sharma M, Burré J. Synaptic vesicle-omics in mice captures signatures of aging and synucleinopathy. Nat Commun 2025; 16:4079. [PMID: 40312501 PMCID: PMC12046008 DOI: 10.1038/s41467-025-59441-7] [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: 08/31/2024] [Accepted: 04/23/2025] [Indexed: 05/03/2025] Open
Abstract
Neurotransmitter release occurs through exocytosis of synaptic vesicles. α-Synuclein's function and dysfunction in Parkinson's disease and other synucleinopathies is thought to be tightly linked to synaptic vesicle binding. Age is the biggest risk factor for synucleinopathy, and ~15% of synaptic vesicle proteins have been linked to central nervous system diseases. Yet, age- and disease-induced changes in synaptic vesicles remain unexplored. Via systematic analysis of synaptic vesicles at the ultrastructural, protein, and lipid levels, we reveal specific changes in synaptic vesicle populations, proteins, and lipids over age in wild-type mice and in α-synuclein knockout mice with and without expression of human α-synuclein. Strikingly, we find several previously undescribed synaptic changes in mice lacking α-synuclein, suggesting that loss of α-synuclein function contributes to synaptic dysfunction. These findings not only provide insights into synaptic vesicle biology and disease mechanisms in synucleinopathy, but also serve as a baseline for further mechanistic exploration of age- and disease-related alterations in synaptic vesicles.
Collapse
Affiliation(s)
- Virginia Gao
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Julita Chlebowicz
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Karlton Gaskin
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Juan A Briano
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Lauren E Komer
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - André Pineda
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Shrey Jhalani
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Saad Ahmad
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Eseosa Uwaifo
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Luca S Black
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jillian E Haller
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Serge Przedborski
- Departments of Neurology, Pathology & Cell Biology and Neuroscience, Columbia University, New York, NY, 10032, USA
| | - Diane A Lane
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, 14853, USA
| | - Manu Sharma
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jacqueline Burré
- Brain and Mind Research Institute, Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
4
|
Chang A, Sia JT, Casper GE, Wood SR, Markham L, Westhoff LL, Saxton AJ, Snow GL, Van Ry PM, Raghu G. Idiopathic pulmonary fibrosis and murine models of pulmonary fibrosis: correlation of decline in lung function. Eur Respir J 2025; 65:2402317. [PMID: 40268504 DOI: 10.1183/13993003.02317-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025]
Affiliation(s)
- Ashley Chang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Jhon Tolentino Sia
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Garrett E Casper
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Seth R Wood
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Lance Markham
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Luke L Westhoff
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Aubrianna J Saxton
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Gregory L Snow
- Department of Statistics, Brigham Young University, Provo, UT, USA
| | - Pam M Van Ry
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
- P.M. Van Ry and G. Raghu contributed equally to this article as lead authors and supervised the work
| | - Ganesh Raghu
- Department of Medicine, University of Washington, Seattle, WA, USA
- P.M. Van Ry and G. Raghu contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
5
|
Aoki Y, Wakamatsu M, Sono N, Xiao W, Ishii E, Nagai T, Nagai Y, Fujiwara Y, Kunieda T, Otsuki J. Impact of aging on spermatogenic function and reproductive outcomes in repro57 heterozygous male mice: A model for age-related infertility. J Assist Reprod Genet 2025:10.1007/s10815-025-03481-x. [PMID: 40257706 DOI: 10.1007/s10815-025-03481-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 04/08/2025] [Indexed: 04/22/2025] Open
Abstract
PURPOSE This study aims to investigate the histological changes, sperm parameters, and their impact on embryo development rates and offspring numbers in advanced-age male repro57 heterozygous mice, corresponding to approximately 40 years of age in humans. METHODS Sperm parameters were assessed in both young and advanced-age repro57 heterozygous mice, as well as in young and advanced-age wild-type mice. Additionally, testis weight and histological analysis of seminiferous tubules were conducted to identify degenerative changes. Male mice from each group were mated with young wild-type females to compare offspring numbers, and in vitro fertilization (IVF) was used to evaluate fertilization and blastocyst formation rates. RESULTS No significant differences in sperm concentration and motility were observed between young and aged wild-type mice or between young wild-type and young repro57 heterozygous mice. However, advanced-age repro57 heterozygous mice exhibited significantly lower sperm parameters and testis weight compared to advanced-age wild-type mice. Histological analysis revealed increased Sertoli cell vacuolation in the seminiferous tubules of advanced-age repro57 heterozygous mice. Additionally, these advanced-age mice exhibited significantly lower blastocyst formation rates and produced fewer offspring compared to advanced-age wild-type mice. CONCLUSION Advanced reproductive aging in repro57 heterozygous male mice is associated with marked senescence-like degenerative changes, leading to a decline in offspring numbers, attributed to increased Sertoli cell vacuolation and diminished sperm quality.
Collapse
Affiliation(s)
- Yuto Aoki
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Misaki Wakamatsu
- Department of Animal Science, Faculty of Agriculture, Okayama University, 1 - 1- 1 Tsushimanaka, Tsushimanaka, KitaKita, OkayamaOkayama, 700 - 8530, Japan
| | - Nanami Sono
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Wei Xiao
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Emi Ishii
- Nagai Mother's Hospital, Kamihikona, Misato, Saitama, 341 - 0004, Japan
| | - Takeshi Nagai
- Nagai Mother's Hospital, Kamihikona, Misato, Saitama, 341 - 0004, Japan
| | - Yasushi Nagai
- Nagai Mother's Hospital, Kamihikona, Misato, Saitama, 341 - 0004, Japan
| | - Yasuhiro Fujiwara
- Laboratory of Pathology and Development, Institute for Quantitative Biosciences, The University of Tokyo, YayoiTokyo, Bunkyo, 113 - 0032, Japan
| | - Tetsuo Kunieda
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan
| | - Junko Otsuki
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan.
- Assisted Reproductive Technology Center, Okayama University, 1 - 1- 1 Tsushimanaka, Kita, Okayama, 700 - 8530, Japan.
| |
Collapse
|
6
|
Xiong C, Yu Z, Yin Y, Zhu Q, Ba R, Shen Y, Li H, Wei Z, Lin Z, Wu D. Longitudinal changes of blood-brain barrier and transcytolemmal water exchange permeability in Alzheimer's disease mice: A non-contrast MRI study. Neuroimage 2025; 310:121141. [PMID: 40089221 DOI: 10.1016/j.neuroimage.2025.121141] [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: 11/25/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025] Open
Abstract
Growing evidence suggests that Alzheimer's disease (AD) has been linked with the dysfunction of glymphatic system. Previous studies were primarily cross-sectional and focused on only one specific component, hindering the understanding of overall glymphatic function in AD. We evaluated the longitudinal changes in multiple components of glymphatic system (blood-brain barrier (BBB) and transcytolemmal water exchange (TWE) permeability) in AD mice. Five female wild-type and four 3 × Tg-AD mice from 5 to 13 months of age were scanned monthly using two non-contrast MRI techniques, water-extraction-with-phase-contrast-arterial-spin-tagging (WEPCAST) and diffusion-time-dependent kurtosis imaging (tDKI), yielding BBB and TWE permeability. Immunostaining was used to evaluate tight junction proteins associated with BBB structural integrity, aquaporin 4 (AQP4) related to TWE, and AQP4 perivascular space (PVS) polarization that might represent PVS-parenchyma water exchange. The relationship between glymphatic function and AD pathology, as measured by amyloid beta (Aβ) and tau deposition, was also explored. Our results revealed significantly increased BBB and hippocampal TWE permeability in AD mouse brains, consistent with the histological findings of reduced tight junction proteins and upregulated AQP4, which were correlated with each other and can be predictive of Aβ and tau deposition. Impaired AQP4 PVS polarization was also found in AD mice. In conclusion, water exchange in multiple components of glymphatic system altered in AD mice, and these in vivo MRI findings were validated pathologically, which might affect the waste clearance in the glymphatic neurofluid.
Collapse
Affiliation(s)
- Chuhan Xiong
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Ziyang Yu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Yu Yin
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Qinfeng Zhu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Ruicheng Ba
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Yao Shen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Haotian Li
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China
| | - Zhiliang Wei
- Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zixuan Lin
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China.
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310027, PR China.
| |
Collapse
|
7
|
Basu L, Grieco-St-Pierre L, Ching MEA, Stead JD, Hanson AA, Palaniyandi J, van Zyl E, Hoyeck MP, McKay KS, van Allen KA, Lee H, Dai XQ, Bautista A, Fadzeyeva E, Mulvihill EE, Yauk CL, Mennigen JA, MacDonald PE, Bruin JE. Cisplatin Exposure Dysregulates Insulin Secretion in Male and Female Mice. Diabetes 2025; 74:528-543. [PMID: 39808439 PMCID: PMC11926276 DOI: 10.2337/db24-0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
ARTICLE HIGHLIGHTS Cancer survivors who receive cisplatin chemotherapy have an increased risk of type 2 diabetes, but the underlying mechanisms remain unclear. The aim of this study was to investigate whether cisplatin impacts β-cell health and function, thereby contributing to increased type 2 diabetes risk in cancer survivors. In vivo and in vitro cisplatin exposure dysregulated insulin secretion in male and female mice. In vitro cisplatin exposure reduced oxygen consumption, impaired β-cell exocytotic capacity, and altered expression of genes within the insulin secretion pathway in mouse islets. Understanding how chemotherapeutic drugs cause β-cell injury is critical for designing targeted interventions to reduce the risk of cancer survivors developing type 2 diabetes after treatment.
Collapse
Affiliation(s)
- Lahari Basu
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Lili Grieco-St-Pierre
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Ma. Enrica Angela Ching
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - John D.H. Stead
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Antonio A. Hanson
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Jana Palaniyandi
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Erin van Zyl
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Myriam P. Hoyeck
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Kelsea S. McKay
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Kyle A. van Allen
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Hyojin Lee
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Xiao-Qing Dai
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Austin Bautista
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Evgenia Fadzeyeva
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Erin E. Mulvihill
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Carole L. Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jan A. Mennigen
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Patrick E. MacDonald
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Jennifer E. Bruin
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
Sunbul FS, Almuqbil RM, Zhang H, Alhudaithi SS, Fernandez ME, Aldaqqa RR, Garcia VA, Robila V, Halquist MS, Gordon SW, Bos PD, da Rocha SRP. An improved experimental model of osteosarcoma lung metastases to investigate innovative therapeutic interventions and sex as a biological variable. Int J Pharm 2025; 673:125372. [PMID: 39971171 DOI: 10.1016/j.ijpharm.2025.125372] [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: 11/15/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy, with OS lung metastasis (OSLM) being the leading cause of death in OS patients. No curative pharmacotherapies for OSLM are available, highlighting the clinical need for new therapies. Improved and rigorous preclinical models of OSLM are key in supporting advancements in this field. We aimed to develop an immunocompetent mouse model of OSLM that allows monitoring pharmacotherapies' effect on the lung metastatic burden over time and assessing the impact of sex as a biological variable in tumor growth and response to therapy. We transformed K7M2 cells to express bioluminescence and fluorescence, enabling real-time tracking of OSLM in BALB/c mice following tail vein injection. Metastasis was confined to the lungs and exhibited exponential growth with typical downregulated Fas receptor expression. In vivo bioluminescence correlated strongly with ex vivo, suggesting its reliability for evaluating metastatic progression and therapy response. Fluorescence from tdT was stable upon tissue processing, providing unique opportunities to probe the tumor characteristics ex vivo. We also assessed the effect of local lung-delivered gemcitabine, which was well-tolerated and significantly reduced OSLM burden without causing pulmonary toxicity. However, treatment did not resolve metastatic disease. We also explored the effect of sex on tumor growth and response to therapy; while no difference was observed in tumor growth between male and female mice, females showed a better response to local gemcitabine administration. In sum, we established a robust and rigorous immunocompetent mouse model of OSLM that will facilitate exploring new pharmacotherapies for OSLM.
Collapse
Affiliation(s)
- Fatemah S Sunbul
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Rashed M Almuqbil
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Hanming Zhang
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sulaiman S Alhudaithi
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew E Fernandez
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Raneem R Aldaqqa
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Victoria A Garcia
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Valentina Robila
- Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Matthew S Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sarah W Gordon
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Thomas JeffersonUniversity, Philadelphia, PA, the United States of America
| | - Paula D Bos
- Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Department of Pathology - School of Medicine, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America
| | - Sandro R P da Rocha
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Center for Pharmaceutical Engineering and Sciences - School of Pharmacy, Virginia Commonwealth University, Richmond, VA, the United States of America; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, the United States of America.
| |
Collapse
|
9
|
Teitz M, Velarde E, Yang X, Lee S, Lecksell K, Terrillion C, Bibic A, Ngen EJ. Developing Magnetic Resonance Imaging Biomarkers of Neuroinflammation, Cognitive Impairment, and Survival Outcomes for Radiotherapy-Induced Brain Injury in a Preclinical Mouse Model. Invest Radiol 2025:00004424-990000000-00306. [PMID: 40095964 DOI: 10.1097/rli.0000000000001173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
OBJECTIVE Radiotherapy-induced brain injury (RIBI) is a chronic side effect that affects up to 90% of brain tumor survivors treated with radiotherapy. Here, we used multiparametric magnetic resonance imaging (MRI) to identify noninvasive and clinically translatable biomarkers of RIBI. METHOD 8-week-old female, immune competent BALB/c mice were stereotactically irradiated with a single dose of 80 Gy, at a dose rate of 1.7 Gy/minute. The irradiated mice were then monitored longitudinally with MRI, behavioral tests of learning and memory, and immunohistochemistry, in comparison to nonirradiated mice. RESULTS Three types of MRI biomarkers of RIBI were identified. A contrast-enhanced T1-weighted MRI biomarker was identified as being best suited to detect the onset of injury, by detecting changes in the blood-brain barrier (BBB) permeability. Maximum BBB permeability (18.95 ± 1.75) was detected with contrast-enhanced T1-weighted MRI at 1-month postirradiation in irradiated mice (P < 0.0001, n = 3). Interestingly, maximum neuroinflammation (24.14 ± 6.72) was also detected using IBA1 and CD68 immunohistochemistry at 1-month postirradiation in irradiated mice (P = 0.0041, n = 3). This simultaneous maximum BBB permeability and neuroinflammation detection also coincided with the detection of the onset of transient cognitive impairment, detected using the fear-conditioning behavioral test at 1-month postirradiation in irradiated mice compared to nonirradiated mice (P = 0.0017, n = 10). A T2-weighted MRI hyperintensity biomarker was also identified, and determined to be best suited to detect intermediate injury. Maximum T2-weighted MRI hyperintensity (3.97 ± 2.07) was detected at 2-month postirradiation in the irradiated mice compared to nonirradiated mice (P = 0.0368, n = 3). This T2-weighted MRI hyperintensity also correlated with maximum astrogliosis (9.92 ± 4.21), which was also detected at 2-month postirradiation using GFAP immunohistochemistry in the irradiated mice compared to nonirradiated mice (P = 0.0215, n = 3). Finally, T2-weighted and T2*-weighted MRI hypointensity biomarkers were identified as being best suited to detect late injury, from 4-month postirradiation. These biomarkers correlated with increased iron deposition from late vascular damage, which was validated with Perls' Prussian blue histology (P < 0.05, n = 3). These hypointense MRI biomarkers of late injury also preceded significant weight loss, severe cognitive impairment, and decreased survival in the irradiated mice compared to the nonirradiated mice. CONCLUSIONS Here, we identified 3 types of translational MRI biomarkers of RIBI that could enable the noninvasive longitudinal evaluation of potential RIBI prophylactic and therapeutic agents. These translational MRI biomarkers could also play a pivotal role in the management of RIBI in brain tumor survivors.
Collapse
Affiliation(s)
- Maya Teitz
- From the Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD (M.T., X.Y., A.B., E.J.N.); Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD (E.V.); Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD (S.L.); Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD (K.L.); Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, (C.T.); F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD (A.B.); and Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD (E.J.N.)
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Takahashi H, Suzuki J, Ikeda R, Hisaoka T, Koshiba Y, Ohta J, Hirano-Kawamoto A, Katori Y. Time-course changes in videofluoroscopic findings during developmental and aging stages in mice. Neuroreport 2025; 36:211-216. [PMID: 39976015 DOI: 10.1097/wnr.0000000000002140] [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] [Indexed: 02/21/2025]
Abstract
Videofluoroscopic swallow study methodology in mice has recently been established; however, details of developmental and age-related changes remain unclear. This study aimed to reveal the changes in swallowing associated with the entire lifespan of mice using the videofluoroscopic swallow study methodology. We tested six age groups of male C57BL/6J mice (4, 8, 16, 54, 77, and 104 weeks old; n = 5 for each group, respectively). The videofluoroscopic swallow study analyzed the bolus area, pharyngeal transit time, interswallow interval, lick-swallow ratio, and lick rate. Significant age differences were found in the bolus area and lick rate. The bolus area increased significantly as the mice grew older. There was a significant increase in the lick rate from 4 to 8 weeks ( P < 0.001) and then a significant decrease from 8 to 104 weeks ( P < 0.01). No significant difference was observed between the various stages in the lick-swallow ratio, interswallow interval, or pharyngeal transit time. There was a significant positive correlation between the body weight and bolus area. This study revealed the detailed changes in swallowing at each life stage from swallowing maturation to old age in the mice. Our research may serve as a guide for future studies on swallowing in humans.
Collapse
Affiliation(s)
- Hiyori Takahashi
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University School of Medicine, Sendai
| | - Jun Suzuki
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University School of Medicine, Sendai
| | - Ryoukichi Ikeda
- Department of Otolaryngology, Head and Neck Surgery, Iwate Medical University School of Medicine, Shiwa, Japan
| | - Takuma Hisaoka
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University School of Medicine, Sendai
| | - Yasutoshi Koshiba
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University School of Medicine, Sendai
| | - Jun Ohta
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University School of Medicine, Sendai
| | - Ai Hirano-Kawamoto
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University School of Medicine, Sendai
| | - Yukio Katori
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University School of Medicine, Sendai
| |
Collapse
|
11
|
Clare AJ, Langer PM, Ward A, Chan YK, Dick AD, Copland DA. Characterization of the ocular inflammatory response to AAV reveals divergence by sex and age. Mol Ther 2025; 33:1246-1263. [PMID: 39825566 PMCID: PMC11897812 DOI: 10.1016/j.ymthe.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/12/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
Progress for ocular adeno-associated virus (AAV) gene therapy has been hindered by AAV-induced inflammation, limiting dose escalation and long-term efficacy. Broadly, the extent of inflammatory responses alters with age and sex, yet these factors are poorly represented in pre-clinical development of ocular AAV gene therapies. Here, we combined clinical imaging, flow cytometry, and bulk sequencing of sorted microglia to interrogate the longitudinal inflammatory response following intravitreal delivery of AAV2 in young (3-month-old), middle aged (9-month-old), and old (18-month-old) Cx3cr1-creER:R26tdTomato+/- mice of both sexes. Young males and females exhibited a similar dynamic response, with peak inflammation evident at days 10-12 and signs of clinical resolution by day 28. However, the magnitude of the transcriptional response by microglia and adaptive T cell infiltrate differed between sexes. With age, increased and persistent inflammation were observed in both sexes, although old males maintained their microglia transcriptional AAV response signature. Contrarily, females demonstrated greater divergence in their inflammatory response across age, with enriched cellular stress and inflammatory gene expression in older mice and corresponding signs of retinal degeneration. These findings inform crucial sex and age differences for the therapeutic application of ocular gene therapy, highlighting the need to further understand these factors to overcome AAV immunogenicity.
Collapse
Affiliation(s)
- Alison J Clare
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, BS8 1TD Bristol, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, EC1V 2PD London, UK.
| | - Philip M Langer
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, BS8 1TD Bristol, UK
| | - Amy Ward
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, BS8 1TD Bristol, UK
| | - Ying Kai Chan
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, BS8 1TD Bristol, UK; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, BS8 1TD Bristol, UK; School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD Bristol, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, EC1V 2PD London, UK; University College London Institute of Ophthalmology, EC1V 9EL London, UK
| | - David A Copland
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, BS8 1TD Bristol, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, EC1V 2PD London, UK.
| |
Collapse
|
12
|
Brandão SR, Lazzari E, Vitorino R, Meroni G, Reis-Mendes A, Neuparth MJ, Amado F, Carvalho F, Ferreira R, Costa VM. Comprehensive ubiquitome analysis reveals persistent mitochondrial remodeling disruptions from doxorubicin-induced cardiotoxicity in aged CD-1 male mice. Arch Toxicol 2025:10.1007/s00204-025-04006-2. [PMID: 40035845 DOI: 10.1007/s00204-025-04006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
Doxorubicin (DOX)-associated cardiotoxicity is characterized by long-term manifestations, whose mechanisms remain incompletely understood, and is exacerbated by various risk factors, with age being a prominent contributor. The objective of this study was to assess the enduring cardiac molecular impacts of DOX in old CD-1 male mice, focusing on ubiquitinated proteins. At 19 months of age, DOX group received a cumulative dose of 9.0 mg/kg of DOX, while control animals got saline solution. Animals were sacrificed 2 months after the administration. DOX induced heart structural changes and increased proteolytic activity. Additionally, increased protein ubiquitination was observed in DOX group, despite the decreased content of the E3 ubiquitin-protein ligase Atrogin-1. A search of poly-ubiquitinated proteins, enriched by tandem ubiquitin-binding entities (TUBEs), showed increased poly-ubiquitination of proteins associated with sarcomere organization and mitochondrial metabolism processes by DOX. Increased mitochondrial density inferred by higher citrate synthase activity was found in DOX group. Moreover, decreased biogenesis and auto(mito)phagy occurred in DOX animals, proven by decreased peroxisome proliferator-activated receptor γ coactivator 1 α, Beclin1 and microtubule-associated protein light chain 3 content. These findings indicate a reduction in mitochondrial biogenesis and accumulation of dysfunctional mitochondria in the aged heart, along with elevated levels of poly-ubiquitinated proteins after DOX treatment. Thus, the disruption of mitochondrial remodeling and impaired protein ubiquitination emerge as enduring consequences of DOX-induced cardiotoxicity, persisting for even 2 months after DOX exposure. This underscores the long-lasting impact of DOX, with significant effects continuing beyond the period of administration, which advocates for longer clinical surveillance.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Elisa Lazzari
- Molecular Genetics Lab, Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Rui Vitorino
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
- Institute of Biomedicine (Ibimed), Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
- Department of Surgery and Physiology, Faculty of Medicine, UnIC@RISE, University of Porto, 4200-319, Porto, Portugal
| | - Germana Meroni
- Molecular Genetics Lab, Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- LAQV-REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Maria João Neuparth
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
| | - Francisco Amado
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
13
|
Rajput M, Malik IA, Methi A, Cortés Silva JA, Fey D, Wirths O, Fischer A, Wilting J, von Arnim CAF. Cognitive decline and neuroinflammation in a mouse model of obesity: An accelerating role of ageing. Brain Behav Immun 2025; 125:226-239. [PMID: 39730092 DOI: 10.1016/j.bbi.2024.12.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/30/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024] Open
Abstract
Obesity, a pandemic, worldwide afflicts almost one billion people. Obesity and ageing share several pathological pathways leading to neurological disorders. However, due to a lack of suitable animal models, the long-term effects of obesity on age-related disorders- cognitive impairment and dementia have not yet been thoroughly investigated. Therefore, the current investigation focuses on developing a suitable model to explore the effects of obese-ageing. It also aims to determine whether obesity affects cognitive abilities in an age-dependent manner, and to identify a potential biomarker(s) for cognitive decline. Cognitive tests were carried out on 6-months and 1-year-old melanocortin-4 receptor (Mc4r)-deficient-obese and lean (wildtype) mice. Additionally, brains and sera were harvested for molecular, histological and serological analyses from 6, 12, and 24-months-old mice. Finally, RT-PCR was carried out after hippocampal mRNA sequencing. The cognitive tests revealed that 1-year-old obese mice have cognitive impairment along with underlying neurodegenerative changes, such as enlarged lateral ventricles. Serum neurofilament light chain (sNfL) levels were also elevated. Lipid accumulation and neuroinflammation were apparent besides, a compromised blood-brain barrier (BBB) indicated by altered junction protein gene expression. Differentially-expressed genes associated with cognitive decline were identified by mRNA sequencing of hippocampi. One such gene, Secreted Phosphoprotein 1 (Spp1) had markedly increased expression in cognitively-impaired obese mice. Our findings present an obese-aged mouse model of cognitive decline with neuroinflammation, reduced BBB-integrity and predisposing neurodegenerative changes. Obese-ageing accelerates the progression of cognitive impairment. Furthermore, Spp1 appears to be a potential biomarker for early diagnosis of neuropathological disorders.
Collapse
Affiliation(s)
- Mansi Rajput
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany.
| | - Ihtzaz Ahmed Malik
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany.
| | - Aditi Methi
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany.
| | - Jonathan Alexis Cortés Silva
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany.
| | - Dorothea Fey
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany.
| | - Oliver Wirths
- Department of Psychiatry, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany.
| | - André Fischer
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Von-Siebold-Str. 3a, 37075 Goettingen, Germany; Department of Psychiatry, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany.
| | - Jörg Wilting
- Institute of Anatomy and Embryology, University Medical Center Goettingen, Kreuzbergring 36, D-37075 Goettingen, Germany.
| | - Christine A F von Arnim
- Department of Geriatrics, University Medical Center Goettingen, Robert-Koch-Str. 42, 37075 Goettingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
14
|
Ramnauth AD, Tippani M, Divecha HR, Papariello AR, Miller RA, Nelson ED, Thompson JR, Pattie EA, Kleinman JE, Maynard KR, Collado-Torres L, Hyde TM, Martinowich K, Hicks SC, Page SC. Spatiotemporal analysis of gene expression in the human dentate gyrus reveals age-associated changes in cellular maturation and neuroinflammation. Cell Rep 2025; 44:115300. [PMID: 40009515 DOI: 10.1016/j.celrep.2025.115300] [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: 05/24/2024] [Revised: 09/19/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
The dentate gyrus of the hippocampus is important for many cognitive functions, including learning, memory, and mood. Here, we present transcriptome-wide spatial gene expression maps of the human dentate gyrus and investigate age-associated changes across the lifespan. Genes associated with neurogenesis and the extracellular matrix are enriched in infants and decline throughout development and maturation. Following infancy, inhibitory neuron markers increase, and cellular proliferation markers decrease. We also identify spatio-molecular signatures that support existing evidence for protracted maturation of granule cells during adulthood and age-associated increases in neuroinflammation-related gene expression. Our findings support the notion that the hippocampal neurogenic niche undergoes major changes following infancy and identify molecular regulators of brain aging in glial- and neuropil-enriched tissue.
Collapse
Affiliation(s)
- Anthony D Ramnauth
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Alexis R Papariello
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Ryan A Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Erik D Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jacqueline R Thompson
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD 21205, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21205, USA; Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
15
|
Lee CW, Wang BYH, Wong SH, Chen YF, Cao Q, Hsiao AWT, Fung SH, Chen YF, Wu HH, Cheng PY, Chou ZH, Lee WYW, Tsui SKW, Lee OKS. Ginkgolide B increases healthspan and lifespan of female mice. NATURE AGING 2025; 5:237-258. [PMID: 39890935 DOI: 10.1038/s43587-024-00802-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 12/20/2024] [Indexed: 02/03/2025]
Abstract
Various anti-aging interventions show promise in extending lifespan, but many are ineffective or even harmful to healthspan. Ginkgolide B (GB), derived from Ginkgo biloba, reduces aging-related morbidities such as osteoporosis, yet its effects on healthspan and longevity have not been fully understood. In this study, we found that continuous oral administration of GB to female mice beginning at 20 months of age extended median survival and median lifespan by 30% and 8.5%, respectively. GB treatment also decreased tumor incidence; enhanced muscle quality, physical performance and metabolism; and reduced systemic inflammation and senescence. Single-nucleus RNA sequencing of skeletal muscle tissue showed that GB ameliorated aging-associated changes in cell type composition, signaling pathways and intercellular communication. GB reduced aging-induced Runx1+ type 2B myonuclei through the upregulation of miR-27b-3p, which suppresses Runx1 expression. Using functional analyses, we found that Runx1 promoted senescence and cell death in muscle cells. Collectively, these findings suggest the translational potential of GB to extend healthspan and lifespan and to promote healthy aging.
Collapse
Affiliation(s)
- Chien-Wei Lee
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan.
- Department of Biomedical Engineering, China Medical University, Taichung, Taiwan.
| | - Belle Yu-Hsuan Wang
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Shing Hei Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yi-Fan Chen
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Qin Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Allen Wei-Ting Hsiao
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sin-Hang Fung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu-Fan Chen
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
- Department of Biomedical Engineering, China Medical University, Taichung, Taiwan
| | - Hao-Hsiang Wu
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Po-Yu Cheng
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Zong-Han Chou
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Wayne Yuk-Wai Lee
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Stephen Kwok Wing Tsui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
16
|
Hong Z, Yi S, Deng M, Zhong Y, Zhao Y, Li L, Zhou H, Xiao Y, Hu X, Niu L. Transcranial Focused Ultrasound Modifies Disease Progression in SOD1G93A Mouse Model of Amyotrophic Lateral Sclerosis. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2025; 72:191-201. [PMID: 40030850 DOI: 10.1109/tuffc.2024.3525143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressively worsening neurodegenerative condition with very few treatment options available. Ultrasound neuromodulation offers promising benefits for treating neurodegenerative diseases such as Parkinson's and Alzheimer's diseases. However, the effects and underlying mechanisms of ultrasound neuromodulation on ALS remain unclear. A head-mounted ultrasound neuromodulation system was developed to noninvasively stimulate the motor cortex of symptomatic mice carrying the G93A human SOD1 mutation (SOD $1^{\text {G93A}}$ ) for four weeks. Motor performance was assessed through the rotarod locomotor test, grip strength test, and open field test. In addition, the effect of ultrasound stimulation on the elastic modulus of gastrocnemius muscle atrophy was measured using real-time shear wave elastography (SWE). Subsequently, the brain tissues of the mice were harvested. Gastrocnemius morphology was examined using hematoxylin-eosin and Gomori aldehyde-fuchsin (GAF) staining. The number of neurons and the phenotype of microglia in the motor cortex were observed by immunohistochemical analysis. Ultrasound therapy delayed disease onset by 10.7% and increased the lifespan by 6.7% in SOD $1^{\text {G93A}}$ mice by reduction of neuronal loss and enhancement of M2 microglia in the motor cortex. Furthermore, we found significant improvements in motor function for ultrasound-treated mice. More importantly, ultrasound stimulation ameliorated gastrocnemius muscle atrophy in the SOD $1^{\text {G93A}}$ mice. These results revealed the neuroprotective effects of ultrasound against the disease pathogenesis of SOD $1^{\text {G93A}}$ mice. Transcranial ultrasound neuromodulation provides an innovative tool for the intervention and treatment of neurodegenerative diseases.
Collapse
|
17
|
Wada Y, Inoko M, Ishihara K, Fukumoto K, Tsurudome Y, Horiguchi M, Fujimura A, Ushijima K. Aging Reduces ATP-Binding Cassette Transporter Expression in Brain Microvessels of Mice. Pharmaceuticals (Basel) 2025; 18:191. [PMID: 40006002 PMCID: PMC11859312 DOI: 10.3390/ph18020191] [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: 12/06/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Background: ATP-binding cassette (ABC) transporters are expressed in the vascular walls of brain capillaries and remove toxic chemicals from the brain. The expression of ABC transporters in peripheral organs is transcriptionally regulated by clock genes and exhibits 24 h periodic fluctuations. In addition, clock gene outputs diminish with aging. In this study, we evaluated whether the expression of ABC transporters in the blood-brain barrier (BBB) of young mice had a 24 h cycle, and whether the expression of ABC transporters in the BBB decreased with age. Methods: Brain microvascular (BMV) fractions from the cerebral cortex of male C57BL/6J mice were prepared using dextran. BMV fractions from young mice (12 weeks old) were prepared every four hours to evaluate 24 h rhythmicity. BMV fractions from both young and aged mice (85 weeks old) were prepared when protein expression peaked (Zeitgeber Time 5). Protein and mRNA expression of ABC transporters in BMV fractions were measured. Results: In young mice, protein expression of P-glycoprotein, breast cancer resistance protein, and multidrug resistance protein 4 showed time-dependent variations with a peak in the light phase (Zeitgeber Time 5); mRNA expression showed no time-dependent variation. The protein expression of these transporters was lower in the BBB of aged mice than in that of young mice, although mRNA expression did not differ between young and aged mice. Conclusions: ABC transporter protein expression levels in BMV endothelial cells decreased with aging; however, mRNA levels did not change, which suggests changes in protein expression did not result from diminished clock gene output. Further studies are needed to elucidate the mechanisms by which ABC transporter expression in the BBB decreases with aging.
Collapse
Affiliation(s)
- Yukiyo Wada
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Masaki Inoko
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Kanako Ishihara
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Karin Fukumoto
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Michiko Horiguchi
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
- Department of Pharmaceutical Engineering, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Akio Fujimura
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
- Division of Clinical Pharmacology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
- Division of Clinical Pharmacology, Jichi Medical University, Tochigi 329-0498, Japan
| |
Collapse
|
18
|
Maupin EA, Adams KL. Cellular Senescence in Glial Cells: Implications for Multiple Sclerosis. J Neurochem 2025; 169:e16301. [PMID: 39831743 PMCID: PMC11745082 DOI: 10.1111/jnc.16301] [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: 09/14/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Aging is the most common risk factor for Multiple Sclerosis (MS) disease progression. Cellular senescence, the irreversible state of cell cycle arrest, is the main driver of aging and has been found to accumulate prematurely in neurodegenerative diseases, including Alzheimer's and Parkinson's disease. Cellular senescence in the central nervous system of MS patients has recently gained attention, with several studies providing evidence that demyelination induces cellular senescence, with common hallmarks of p16INK4A and p21 expression, oxidative stress, and senescence-associated secreted factors. Here we discuss the current evidence of cellular senescence in animal models of MS and different glial populations in the central nervous system, highlighting the major gaps in the field that still remain. As premature senescence in MS may exacerbate demyelination and inflammation, resulting in inhibition of myelin repair, it is critical to increase understanding of cellular senescence in vivo, the functional effects of senescence on glial cells, and the impact of removing senescent cells on remyelination and MS. This emerging field holds promise for opening new avenues of treatment for MS patients.
Collapse
Affiliation(s)
- Elizabeth A. Maupin
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
| | - Katrina L. Adams
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
- The Center for Stem Cells and Regenerative MedicineUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
19
|
Wells AT, Shen MM, Binrouf RH, D'Amico AE, Bossardi Ramos R, Lennartz MR. Identification of Myeloid Protein Kinase C - Epsilon as a Novel Atheroprotective Gene. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.09.627650. [PMID: 39713428 PMCID: PMC11661236 DOI: 10.1101/2024.12.09.627650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Background Atherosclerosis is a lipid mediated chronic inflammatory disease driven my macrophages (MØ). Protein Kinase C - epsilon (PKCɛ) is is a serine/threonine kinase involved in diverse cellular processes such as migration, growth, differentiation, and survival. PKCɛ is known to act in a context dependent manner within heart, however, its role in atherosclerosis is unknown. Methods Bone marrow derived MØ from global PKCɛ KO mice were examined for impact of lipid metabolism and inflammatory factor secretion. Public geneset analysis assessed raw counts of PKCɛ to determine translational relevance. To determine the function myeloid PKCɛ on atherosclerosis a novel murine model was generated using LysM Cre technology. After its characterization, human-like hypercholesterolemia was induced to assess plaque morphology in WT mice or mice lacking myeloid PKCɛ. Results Public geneset analysis of human atherosclerotic plaque tissue revealed that PKCɛ expression is inversely correlated with plaque size and vulnerability. Similarly, peritoneal MØ from hypercholesterolemic mice have significantly lower PKCɛ expression. As MØ play a major role in atherogenesis, we generated a mouse strain with PKCɛ selectively deleted in the myeloid lineage (mɛKO). qPCR revealed no basal differences between genotypes in the expression of lipid uptake receptors, efflux transporters, or inflammatory markers. However, upon lipid loading, mɛKO MØs retained significantly more cholesterol than WT. Human-like hypercholesterolemia was induced in WT and mɛKO mice and assessed for lesion area and plaque morphology in aortic arches and aortic roots. We found that, compared to WT, the lesion area in mɛKO mice was significantly larger, more necrotic, had larger foam cells, and thinner collagen caps. Conclusions Loss of myeloid PKCɛ promotes atherosclerosis as determined by larger lesions, more necrosis, thinner plaque caps). Together, these data identify myeloid PKCɛ as a novel atheroprotective gene, laying the foundation for mechanistic studies on the signaling networks responsible for the phenotype. Highlights A novel murine model in which PKCɛ is floxed (PKCɛ fl/fl ) on both alleles haas been generated, backcrossed, and deposited into Jackson Laboratories. PKCε fl/fl mice have been crossed with those on the LysM Cre background thereby deleting PKCε from myeloid cells (mεKO). Deletion of PKCε has no basal affects on other PKC isoforms, lipid handling markers, or inflammatory markers.Upon stimulation with lopid loading in vitro or hypercholesterolemia in vivo, mεKO BMDMs retain more cholesterol and mεKO mice develop a more vulnerable plaque phenotype (i.e. larger lesions, more necrosis, thimmer plaque caps).These findings provide a rationale for the need to identify mediators in the PKCε signaling pathway responsible for protection against vulnerable plaques in atherosclerosis; potentially aiding in the development of preventative and therapeutic treatments.
Collapse
|
20
|
Tao C, Lin S, Shi Y, Gong W, Chen M, Li J, Zhang P, Yao Q, Qian D, Ling Z, Xiao G. Inactivation of Tnf-α/Tnfr signaling attenuates progression of intervertebral disc degeneration in mice. JOR Spine 2024; 7:e70006. [PMID: 39391171 PMCID: PMC11461905 DOI: 10.1002/jsp2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
Background Intervertebral disc degeneration (IVDD) is a major cause of low back pain (LBP), worsened by chronic inflammatory processes associated with aging. Tumor necrosis factor alpha (Tnf-α) and its receptors, Tnf receptor type 1 (Tnfr1) and Tnf receptor type 2 (Tnfr2), are upregulated in IVDD. However, its pathologic mechanisms remain poorly defined. Methods To investigate the role of Tnfr in IVDD, we generated global Tnfr1/2 double knockout (KO) mice and age-matched control C57BL/6 male mice, and analyzed intervertebral disc (IVD)-related phenotypes of both genotypes under physiological conditions, aging, and lumbar spine instability (LSI) model through histological and immunofluorescence analyses and μCT imaging. Expression levels of key extracellular matrix (ECM) proteins in aged and LSI mice, especially markers of cell proliferation and apoptosis, were evaluated in aged (21-month-old) mice. Results At 4 months, KO and control mice showed no marked differences of IVDD-related parameters. However, at 21 months of age, the loss of Tnfr expression significantly alleviated IVDD-like phenotypes, including a significant increase in height of the nucleus pulposus (NPs) and reductions of endplates (EPs) porosity and histopathological scores, when compared to controls. Tnfr deficiency promoted anabolic metabolism of the ECM proteins and suppressed ECM catabolism. Tnfr loss largely inhibited hypertrophic differentiation, and, in the meantime, suppressed cell apoptosis and cellular senescence in the annulus fibrosis, NP, and EP tissues without affecting cell proliferation. Similar results were observed in the LSI model, where Tnfr deficiency significantly alleviated IVDD and enhanced ECM anabolic metabolism while suppressing catabolism. Conclusion The deletion of Tnfr mitigates age-related and LSI-induced IVDD, as evidenced by preserved IVD structure, and improved ECM integrity. These findings suggest a crucial role of Tnf-α/Tnfr signaling in IVDD pathogenesis in mice. Targeting this pathway may be a novel strategy for IVDD prevention and treatment.
Collapse
Affiliation(s)
- Chu Tao
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbinChina
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
| | - Sixiong Lin
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
- Department of OrthopaedicsThe First Affiliated Hospital of Guangzhou Medical University, Guangdong key Laboratory of Orthopaedic Technology and Implant MaterialsGuangzhouChina
| | - Yujia Shi
- School of Biomedical SciencesThe Chinese University of Hong KongShatinHong Kong
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung HomHong Kong
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
| | - Jianglong Li
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
- Department of Orthopaedics, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Peijun Zhang
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
| | - Dongyang Qian
- Department of OrthopaedicsThe First Affiliated Hospital of Guangzhou Medical University, Guangdong key Laboratory of Orthopaedic Technology and Implant MaterialsGuangzhouChina
| | - Zemin Ling
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopaedic SurgeryThe Seventh Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease ResearchSouthern University of Science and TechnologyShenzhenChina
| |
Collapse
|
21
|
Horstmann H, Michel NA, Sheng X, Hansen S, Lindau A, Pfeil K, Fernández MC, Marchini T, Winkels H, Mitre LS, Abogunloko T, Li X, Mwinyella TBN, Gissler MC, Bugger H, Heidt T, Buscher K, Hilgendorf I, Stachon P, Piepenburg S, Verheyen N, Rathner T, Gerhardt T, Siegel PM, Oswald WK, Cohnert T, Zernecke A, Madl J, Kohl P, Foks AC, von zur Muehlen C, Westermann D, Zirlik A, Wolf D. Cross-species single-cell RNA sequencing reveals divergent phenotypes and activation states of adaptive immunity in human carotid and experimental murine atherosclerosis. Cardiovasc Res 2024; 120:1713-1726. [PMID: 39041203 PMCID: PMC11587564 DOI: 10.1093/cvr/cvae154] [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: 11/16/2023] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024] Open
Abstract
AIMS The distinct functions of immune cells in atherosclerosis have been mostly defined by pre-clinical mouse studies. Contrastingly, the immune cell composition of human atherosclerotic plaques and their contribution to disease progression are only poorly understood. It remains uncertain whether genetic animal models allow for valuable translational approaches. METHODS AND RESULTS Single-cell RNA-sequencing (scRNA-seq) was performed to define the immune cell landscape in human carotid atherosclerotic plaques. The human immune cell repertoire demonstrated an unexpectedly high heterogeneity and was dominated by cells of the T-cell lineage, a finding confirmed by immunohistochemistry. Bioinformatical integration with 7 mouse scRNA-seq data sets from adventitial and atherosclerotic vascular tissue revealed a total of 51 identities of cell types and differentiation states, of which some were only poorly conserved between species and exclusively found in humans. Locations, frequencies, and transcriptional programmes of immune cells in mouse models did not resemble the immune cell landscape in human carotid atherosclerosis. In contrast to standard mouse models of atherosclerosis, human plaque leucocytes were dominated by several T-cell phenotypes with transcriptional hallmarks of T-cell activation and memory formation, T-cell receptor, and pro-inflammatory signalling. Only mice at the age of 22 months partially resembled the activated T-cell phenotype. In a validation cohort of 43 patients undergoing carotid endarterectomy, the abundance of activated immune cell subsets in the plaque defined by multi-colour flow cytometry associated with the extent of clinical atherosclerosis. CONCLUSION Integrative scRNA-seq reveals a substantial difference in the immune cell composition of murine and human carotid atherosclerosis-a finding that questions the translational value of standard mouse models for adaptive immune cell studies. Clinical associations suggest a specific role for T-cell driven (auto-)immunity in human plaque formation and instability.
Collapse
Affiliation(s)
- Hauke Horstmann
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Division of Cardiology, Department of Medicine, NYU Cardiovascular Research Center, NYU Grossmann School of Medicine, 10016 New York, NY, USA
| | - Nathaly Anto Michel
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Xia Sheng
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sophie Hansen
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexandra Lindau
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Katharina Pfeil
- Division of Cardiology, Department of Medicine, NYU Cardiovascular Research Center, NYU Grossmann School of Medicine, 10016 New York, NY, USA
| | - Marbely C Fernández
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Timoteo Marchini
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50923 Cologne, Germany
| | - Lucia Sol Mitre
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79106 Freiburg, Germany
| | - Tijani Abogunloko
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79106 Freiburg, Germany
| | - Xiaowei Li
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Timothy Bon-Nawul Mwinyella
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Mark Colin Gissler
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Heiko Bugger
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Timo Heidt
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Konrad Buscher
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine, University Hospital of Münster, 48149 Münster, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Peter Stachon
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sven Piepenburg
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nicolas Verheyen
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Thomas Rathner
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Teresa Gerhardt
- Department of Cardiology, Angiology and Intensive Care Medicine CBF, Deutsches Herzzentrum der Charité, and Berlin Institute of Health (BIH), 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin , 13353 Berlin, Germany
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, 10029 New York, NY, USA
| | - Patrick Malcolm Siegel
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | | | - Tina Cohnert
- Department of Vascular Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Josef Madl
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Peter Kohl
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Amanda C Foks
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, 2333 CC Leiden, The Netherlands
| | - Constantin von zur Muehlen
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Andreas Zirlik
- Department of Cardiology, University Heart Center Graz, Medical University of Graz, 8036 Graz, Austria
| | - Dennis Wolf
- Department of Cardiology and Angiology I, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
22
|
Lu X, Xiong W, Chen Z, Li Y, Xu F, Yang X, Long M, Guo W, Wu S, Sun L, Wang G. Exercise-conditioned plasma ameliorates postoperative cognitive dysfunction by activating hippocampal cholinergic circuit and enhancing BDNF/TrkB signaling. Cell Commun Signal 2024; 22:551. [PMID: 39558340 PMCID: PMC11572510 DOI: 10.1186/s12964-024-01938-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/10/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a prevalent complication following anesthesia and surgery, particularly in the elderly, leading to increased mortality and reduced quality of life. Despite its prevalence, there are no effective clinical treatments. Exercise has shown cognitive benefits in aging and various diseases, which can be transferred to sedentary animals through plasma. However, it is unclear if exercise-conditioned plasma can replicate these benefits in the context of POCD. METHODS Sixteen-month-old male C57BL/6J mice underwent 30 days of voluntary running wheel training or received systemic administration of exercise-conditioned plasma, followed by tibial fracture surgery under general anesthesia at 17 months of age. Cognitive performance, hippocampal synaptic deficits, neuroinflammation, BDNF/TrkB signaling, and medial septum (MS)-hippocampal cholinergic activity were evaluated through immunohistochemical staining, transmission electron microscopy, Western blotting, and biochemical assays. To investigate the role of hippocampal BDNF signaling and cholinergic activity in the therapeutic effects, the TrkB antagonist ANA-12 and the cholinergic receptor muscarinic 1 (CHRM1) antagonist trihexyphenidyl (THP) were administered via intraperitoneal injection, and adeno-associated virus (AAV) vectors expressing Chrm1 shRNA were delivered via intrahippocampal stereotaxic microinjection. RESULTS Exercise-conditioned plasma mimicked the benefits of exercise, alleviating cognitive decline induced by anesthesia/surgery, restoring hippocampal synapse formation and levels of regulators for synaptic plasticity, inhibiting neuroinflammatory responses to surgery by microglia and astrocytes, augmenting BDNF production and TrkB phosphorylation in hippocampal neurons, astrocytes, and microglia, upregulating MS expression of choline acetyltransferase (CHAT) and hippocampal expression of CHRM1 in neurons and astrocytes, and enhancing hippocampal cholinergic innervation and acetylcholine release. Conversely, ANA-12 administration blocked TrkB activation and reduced the protective effects on cognition, synaptic deficits, and neuroinflammatory reactivity of glial cells post-surgery. Similarly, THP administration or intrahippocampal delivery of AAV-Chrm1 shRNA inhibited the activation of the hippocampal cholinergic circuit by exercise plasma, negating the cognitive and neuropathological benefits and reducing BDNF/TrkB signaling enhancements. CONCLUSION Exercise-conditioned plasma can replicate the protective effects of exercise against anesthesia/surgery-induced neuroinflammation, synaptic, and cognitive impairments, at least partly, through CHRM1-dependent regulation of hippocampal cholinergic activity and BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Xiaodi Lu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Weijie Xiong
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Zhuo Chen
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yurou Li
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Fengyan Xu
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Xue Yang
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Meiwen Long
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Wenhan Guo
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Shuliang Wu
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China.
| | - Liang Sun
- Department of Human Anatomy, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China.
| | - Guonian Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
23
|
Moreira-Pais A, Ferreira R, Baltazar T, Neuparth MJ, Vitorino R, Reis-Mendes A, Costa VM, Oliveira PA, Duarte JA. Long-term effects of the chronic administration of doxorubicin on aged skeletal muscle: An exploratory study in mice. Biochem Biophys Res Commun 2024; 733:150650. [PMID: 39255618 DOI: 10.1016/j.bbrc.2024.150650] [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: 07/02/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
The widely used chemotherapeutic drug doxorubicin (DOX) has been associated with adverse effects on the skeletal muscle, which can persist for years after the end of the treatment. These adverse effects may be exacerbated in older patients, whose skeletal muscle might already be impaired by aging. Nonetheless, the mediators responsible for DOX-induced myotoxicity are still largely unidentified, particularly the ones involved in the long-term effects that negatively affect the quality of life of the patients. Therefore, this study aimed to investigate the long-term effects of the chronic administration of DOX on the soleus muscle of aged mice. For that and to mimic the clinical regimen, a dose of 1.5 mg kg-1 of DOX was administered two times per week for three consecutive weeks in a cumulative dose of 9 mg kg-1 to 19-month-old male mice, which were sacrificed two months after the last administration. Body wasting and the atrophy of the soleus muscle, as measured by a decrease in the cross-sectional area of the soleus muscle fibers, were identified as long-term effects of DOX administration. The atrophy observed was correlated with increased reactive oxygen species production and caspase-3 activity. An impaired skeletal muscle regeneration was also suggested due to the correlation between satellite cells activation and the soleus muscle fibers atrophy. Systemic inflammation, skeletal muscle energy metabolism and neuromuscular junction-related markers do not appear to be involved in the long-term DOX-induced skeletal muscle atrophy. The data provided by this study shed light on the mediators involved in the overlooked long-term DOX-induced myotoxicity, paving the way to the improvement of the quality of life and survival rates of older cancer patients.
Collapse
Affiliation(s)
- Alexandra Moreira-Pais
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto (FADEUP) and Laboratory for Integrative and Translational Research in Population Health (ITR), 4200-450, Porto, Portugal; LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal; Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Telmo Baltazar
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Maria João Neuparth
- Research Center in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto (FADEUP) and Laboratory for Integrative and Translational Research in Population Health (ITR), 4200-450, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal.
| | - Rui Vitorino
- iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - Paula A Oliveira
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.
| | - José A Duarte
- UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, 4585-116, Gandra, Portugal.
| |
Collapse
|
24
|
Zhou Q, Harding JC, Fan P, Spasojevic I, Kovacs A, Akk A, Mitchell A, Springer LE, Gaut JP, Rauch DA, Wickline SA, Pham CTN, Fuh K, Pan H. Safety Evaluations of Rapamycin Perfluorocarbon Nanoparticles in Ovarian Tumor-Bearing Mice. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1752. [PMID: 39513832 PMCID: PMC11547995 DOI: 10.3390/nano14211752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Nanomedicine holds great potential for revolutionizing medical treatment. Ongoing research and advancements in nanotechnology are continuously expanding the possibilities, promising significant advancements in healthcare. To fully harness the potential of nanotechnology in medical applications, it is crucial to conduct safety evaluations for the nanomedicines that offer effective benefits in the preclinical stage. Our recent efficacy studies indicated that rapamycin perfluorocarbon (PFC) nanoparticles showed promise in mitigating cisplatin-induced acute kidney injury (AKI). As cisplatin is routinely administered to ovarian cancer patients as their first-line chemotherapy, in this study, we focused on evaluating the safety of rapamycin PFC nanoparticles in mice bearing ovarian tumor xenografts. Specifically, this study evaluated the effects of repeat-dose rapamycin PFC nanoparticle treatment on vital organs, the immune system, and tumor growth and assessed pharmacokinetics and biodistribution. Our results indicated that rapamycin PFC nanoparticle treatment did not cause any detectable adverse effects on cardiac, renal, or hepatic functions or on splenocyte populations, but it reduced the splenocyte secretion of IL-10, TNFα, and IL12p70 upon IgM stimulation. The pharmacokinetics and biodistribution results revealed a significant enhancement in the delivery of rapamycin to tumors by rapamycin PFC nanoparticles, which, in turn, led to a significant reduction in ovarian tumor growth. Therefore, rapamycin PFC nanoparticles have the potential to be clinically beneficial in cisplatin-treated ovarian cancer patients.
Collapse
Affiliation(s)
- Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - John C. Harding
- Molecular Oncology, Oncology Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ping Fan
- Medical Oncology, Department of Medicine, Duke University, Durham, NC 27708, USA
| | - Ivan Spasojevic
- Medical Oncology, Department of Medicine, Duke University, Durham, NC 27708, USA
| | - Attila Kovacs
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Antonina Akk
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Adam Mitchell
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Luke E. Springer
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph P. Gaut
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel A. Rauch
- Molecular Oncology, Oncology Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel A. Wickline
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- John Cochran Veterans Affairs Medical Center, St. Louis, MO 63106, USA
| | - Katherine Fuh
- Division of Gynecologic Oncology, University of California, San Francisco, NC 90095, USA
| | - Hua Pan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
25
|
Kelberman MA, Rodberg E, Arabzadeh E, Bair-Marshall CJ, Berridge CW, Berrocoso E, Breton-Provencher V, Chandler DJ, Che A, Davy O, Devilbiss DM, Downs AM, Drummond G, Dvorkin R, Fazlali Z, Froemke RC, Glennon E, Gold JI, Ito H, Jiang X, Johansen JP, Kaye AP, Kim JR, Kuo CC, Liu RJ, Liu Y, Llorca-Torralba M, McCall JG, McElligott ZA, McKinney AM, Miguelez C, Min MY, Nowlan AC, Omrani M, Poe GR, Pickering AE, Ranjbar-Slamloo Y, Razquin J, Rodenkirch C, Sales AC, Satyasambit R, Shea SD, Sur M, Tkaczynski JA, Torres-Sanchez S, Uematsu A, Vazquez CR, Vreven A, Wang Q, Waterhouse BD, Yang HW, Yang JH, Zhao L, Zouridis IS, Weinshenker D, Vazey E, Totah NK. Diversity of ancestral brainstem noradrenergic neurons across species and multiple biological factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618224. [PMID: 39464004 PMCID: PMC11507722 DOI: 10.1101/2024.10.14.618224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The brainstem region, locus coeruleus (LC), has been remarkably conserved across vertebrates. Evolution has woven the LC into wide-ranging neural circuits that influence functions as broad as autonomic systems, the stress response, nociception, sleep, and high-level cognition among others. Given this conservation, there is a strong possibility that LC activity is inherently similar across species, and furthermore that age, sex, and brain state influence LC activity similarly across species. The degree to which LC activity is homogenous across these factors, however, has never been assessed due to the small sample size of individual studies. Here, we pool data from 20 laboratories (1,855 neurons) and show diversity across both intrinsic and extrinsic factors such as species, age, sex and brain state. We use a negative binomial regression model to compare activity from male monkeys, and rats and mice of both sexes that were recorded across brain states from brain slices ex vivo or under different anesthetics or during wakefulness in vivo. LC activity differed due to complex interactions of species, sex, and brain state. The LC became more active during aging, independent of sex. Finally, in contrast to the foundational principle that all species express two distinct LC firing modes ("tonic" or "phasic"), we discovered great diversity within spontaneous LC firing patterns. Different factors were associated with higher incidence of some firing modes. We conclude that the activity of the evolutionarily-ancient LC is not conserved. Inherent differences due to age and species-sex-brain state interactions have implications for understanding the role of LC in species-specific naturalistic behavior, as well as in psychiatric disorders, cardiovascular disease, immunology, and metabolic disorders.
Collapse
Affiliation(s)
- Michael A. Kelberman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Ellen Rodberg
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Ehsan Arabzadeh
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, AUS
| | - Chloe J. Bair-Marshall
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Craig W. Berridge
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Esther Berrocoso
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Alicia Che
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Oscar Davy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | | | - Anthony M. Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle Drummond
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roman Dvorkin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Zeinab Fazlali
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- Department of Psychiatry, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Robert C. Froemke
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Otolaryngology, NYU Grossman School of Medicine, New York, NY, USA
| | - Erin Glennon
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Neurology, Weill Cornell Medicine, New York
| | - Joshua I. Gold
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Hiroki Ito
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Xiaolong Jiang
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
- Department of Ophthalmology, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | | | - Alfred P. Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, USA
| | - Jenny R. Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Rong-Jian Liu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yang Liu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordan G. McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew M. McKinney
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Alexandra C. Nowlan
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohsen Omrani
- Department of Psychiatry, Queen’s University, Kingston, ON, Canada
| | - Gina R. Poe
- Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Anthony Edward Pickering
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Yadollah Ranjbar-Slamloo
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Jone Razquin
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Charles Rodenkirch
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Anna C. Sales
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Rath Satyasambit
- RIKEN Center for Brain Science, Wako-shi Saitama, Japan
- Department of Computer Science, Tokyo Institute of Technology, Midori, Yokohama, Japan
| | | | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Sonia Torres-Sanchez
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Akira Uematsu
- Human Informatics and Information Research Institute, National Institute of Advanced Industrial Science and Technology, Japan
| | - Chayla R. Vazquez
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Amelien Vreven
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Hsiu-Wen Yang
- Department of Biomedical Sciences, Chung-Shan Medical University, Taichung, Taiwan
| | - Jen-Hau Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Ioannis S. Zouridis
- Graduate Training Centre of Neuroscience, International Max Planck Research School (IMPRS), University of Tübingen, Tübingen, Germany
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | | | - Elena Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Nelson K. Totah
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| |
Collapse
|
26
|
Kim HY, Son Y, Jeong YJ, Lee SH, Kim N, Ahn YH, Jeon SB, Choi HD, Lee HJ. Effects of 4G Long-Term Evolution Electromagnetic Fields on Thyroid Hormone Dysfunction and Behavioral Changes in Adolescent Male Mice. Int J Mol Sci 2024; 25:10875. [PMID: 39456657 PMCID: PMC11507962 DOI: 10.3390/ijms252010875] [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: 08/22/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Radiofrequency electromagnetic fields (RF-EMFs) can penetrate tissues and potentially influence endocrine and brain development. Despite increased mobile phone use among children and adolescents, the long-term effects of RF-EMF exposure on brain and endocrine development remain unclear. This study investigated the effects of long-term evolution band (LTE) EMF exposure on thyroid hormone levels, crucial for metabolism, growth, and development. Four-week-old male mice (C57BL/6) were exposed to LTE EMF (whole-body average specific absorption rate [SAR] 4 W/kg) or a positive control (lead; Pb, 300 ppm in drinking water) for 4 weeks. Subsequently, the mice underwent behavioral tests including open field, marble burying, and nest building. Blood pituitary and thyroid hormone levels, and thyroid hormone-regulating genes within the hypothalamus-pituitary-thyroid (HPT) axis were analyzed. LTE exposure increased T3 levels, while Pb exposure elevated T3 and T4 and decreased ACTH levels. The LTE EMF group showed no gene expression alterations in the thyroid and pituitary glands, but hypothalamic Dio2 and Dio3 expressions were significantly reduced compared to that in the sham-exposed group. Pb exposure altered the hypothalamic mRNA levels of Oatp1c1 and Trh, pituitary mRNA of Trhr, and Tpo and Tg expression in the thyroid. In conclusion, LTE EMF exposure altered hypothalamic Dio2 and Dio3 expression, potentially impacting the HPT axis function. Further research is needed to explore RF-EMF's impacts on the endocrine system.
Collapse
Affiliation(s)
- Hyun-Yong Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul 01812, Republic of Korea; (H.-Y.K.); (Y.S.); (Y.J.J.); (S.-H.L.)
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju 28160, Republic of Korea
| | - Yeonghoon Son
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul 01812, Republic of Korea; (H.-Y.K.); (Y.S.); (Y.J.J.); (S.-H.L.)
| | - Ye Ji Jeong
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul 01812, Republic of Korea; (H.-Y.K.); (Y.S.); (Y.J.J.); (S.-H.L.)
| | - Soo-Ho Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul 01812, Republic of Korea; (H.-Y.K.); (Y.S.); (Y.J.J.); (S.-H.L.)
| | - Nam Kim
- School of Electrical and Computer Engineering, Chungbuk National University, Cheongju 28466, Republic of Korea;
| | - Young Hwan Ahn
- Department of Neurosurgery, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Sang Bong Jeon
- Department of EMF Research Team, Electronics and Telecommunication Research Institute, Daejon 34129, Republic of Korea;
| | - Hyung-Do Choi
- Department of EMF Research Team, Electronics and Telecommunication Research Institute, Daejon 34129, Republic of Korea;
| | - Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul 01812, Republic of Korea; (H.-Y.K.); (Y.S.); (Y.J.J.); (S.-H.L.)
| |
Collapse
|
27
|
Brandão SR, Oliveira PF, Guerra-Carvalho B, Reis-Mendes A, Neuparth MJ, Carvalho F, Ferreira R, Costa VM. Enduring metabolic modulation in the cardiac tissue of elderly CD-1 mice two months post mitoxantrone treatment. Free Radic Biol Med 2024; 223:199-211. [PMID: 39059512 DOI: 10.1016/j.freeradbiomed.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Mitoxantrone (MTX) is a therapeutic agent used in the treatment of solid tumors and multiple sclerosis, recognized for its cardiotoxicity, with underlying molecular mechanisms not fully disclosed. The cardiotoxicity is influenced by risk factors, including age. Our study intended to assess the molecular effect of MTX on the cardiac muscle of old male CD-1 mice. Mice aged 19 months received a total cumulative dose of 4.5 mg/kg of MTX (MTX group) or saline solution (CTRL group). Two months post treatment, blood was collected, animals sacrificed, and the heart removed. MTX caused structural cardiac changes, which were accompanied by extracellular matrix remodeling, as indicated by the increased ratio between matrix metallopeptidase 2 and metalloproteinase inhibitor 2. At the metabolic level, decreased glycerol levels were found, together with a trend towards increased content of the electron transfer flavoprotein dehydrogenase. In contrast, lower glycolysis, given by the decreased content of glucose transporter GLUT4 and phosphofructokinase, seemed to occur. The findings suggest higher reliance on fatty acids oxidation, despite no major remodeling occurring at the mitochondrial level. Furthermore, the levels of glutamine and other amino acids (although to a lesser extent) were decreased, which aligns with decreased content of the E3 ubiquitin-protein ligase Atrogin-1, suggesting a decrease in proteolysis. As far as we know, this was the first study made in old mice with a clinically relevant dose of MTX, evaluating its long-term cardiac effects. Even two months after MTX exposure, changes in metabolic fingerprint occurred, highlighting enduring cardiac effects that may require clinical vigilance.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Pedro Fontes Oliveira
- LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Bárbara Guerra-Carvalho
- LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal.
| | - Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Maria João Neuparth
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal.
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Rita Ferreira
- LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
28
|
Skurikhin E, Zhukova M, Ermakova N, Pan E, Widera D, Sandrikina L, Kogai L, Kushlinskii N, Kubatiev A, Morozov S, Dygai A. Age-related features of lung cancer treatment using reprogrammed CD8 positive T cells in mice subjected to injection of Lewis lung carcinoma cells. Thorac Cancer 2024; 15:2000-2020. [PMID: 39169897 PMCID: PMC11444928 DOI: 10.1111/1759-7714.15426] [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: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Awareness of age-related features of carcinogenesis and the importance of cellular immunity is crucial for developing effective antitumor therapies for specific patient groups. METHODS In this study, we examined different populations of cancer stem cells (CSCs) and circulating tumor cells (CTCs) in "young" (8-10 weeks) and "aged" (80-82 weeks) C57BL/6 male mice. We used an orthotopic model of Lewis lung carcinoma (LLC) to evaluate the effectiveness of cell therapy targeting lung cancer through reprogrammed CD8-positive T cells (rCD8+ T cells) in mice from two different ages. RESULTS The findings revealed that tumor progression with age is primarily caused by impaired recruitment of T cells to the lungs. Additionally, a lower number of CTCs and CSCs were observed in younger mice compared to the older mice. The antitumor effect of rCD8+ T cells in aged mice was found to be inferior to that in young mice, which can be attributed to the reduced impact of therapy on specific CSCs populations. CONCLUSIONS These results offer new insights into the treatment of lung cancer using rCD8+ T cells. Considering the age-related characteristics influencing disease progression, this therapy has the potential to significantly enhance the effectiveness of treatment methods.
Collapse
Affiliation(s)
| | - Mariia Zhukova
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Natalia Ermakova
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Edgar Pan
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, Whiteknights Campus, Reading, UK
| | - Lubov Sandrikina
- Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Lena Kogai
- Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
- Ministry of Health of the Russian Federation, Siberian State Medical University, Tomsk, Russia
| | | | - Aslan Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Dygai
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
29
|
Majkutewicz I, Kurowska-Rucińska E, Ruciński J, Myślińska D, Grembecka B, Mantej J, Dzik KP. Diverse Efficacy of Dimethyl Fumarate in Alleviating the Late Streptozotocin-Induced Cognitive Impairment and Neuropathological Features in Rat. Mol Neurobiol 2024; 61:7751-7766. [PMID: 38430351 DOI: 10.1007/s12035-024-04024-8] [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: 10/11/2023] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
Our previous study showed that dimethyl fumarate (DMF) treatment performed within three weeks after intracerebroventricular (ICV) injection of streptozotocin (STZ) attenuated spatial memory impairment, hippocampal neurodegeneration, and neuroinflammation in rats. The present study is aimed at verifying the hypothesis that DMF alleviates late effects of STZ (6 months after ICV injection) which reflects advanced stage of the Alzheimer's disease (AD) in human patients. Spatial memory was assessed with Morris water maze (MWM), general brain level of amyloid β (Aβ) and p-tau was measured by western blot, immunofluorescent labelling of active microglia (IBA1), Aβ and p-tau and histological assay of neurodegeneration (Fluoro-Jade C) were performed in hippocampus and cortex. Two-week oral therapy with DMF normalized spatial memory disrupted by STZ but had no influence on general brain level of Aβ and p-tau. However, immunofluorescence showed local reduction of Aβ aggregates number in parietal cortex and p-tau+ cells in CA2 hippocampal area. Microgliosis was alleviated by DMF in CA1 area and parietal cortex. DMF-treated STZ injected rats showed higher number of Aβ containing microglia than untreated group in CA2 and frontal cortex, which may be the result of increased phagocytic activity in these areas after DMF treatment. STZ-induced neurodegeneration was alleviated by DMF in dentate gyrus and frontal cortex. In conclusion DMF treatment exerts beneficial effect on spatial memory in the rat model of late stage of AD, but weakly influences neuropathological features, as only local reduction in number of Aβ aggregates, p-tau containing cells, neurodegeneration, and microgliosis was found.
Collapse
Affiliation(s)
- Irena Majkutewicz
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland.
| | | | - Jan Ruciński
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Dorota Myślińska
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Beata Grembecka
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Jagoda Mantej
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | - Katarzyna P Dzik
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
30
|
Hoshino T, Takase H, Hamanaka G, Kimura S, Fukuda N, Mandeville ET, Lok J, Lo EH, Arai K. Transcriptomic changes in oligodendrocyte lineage cells during the juvenile to adult transition in the mouse corpus callosum. Sci Rep 2024; 14:22334. [PMID: 39333617 PMCID: PMC11436962 DOI: 10.1038/s41598-024-72311-4] [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: 04/12/2024] [Accepted: 09/05/2024] [Indexed: 09/29/2024] Open
Abstract
The corpus callosum, a major white matter tract in the brain, undergoes age-related functional changes. To extend our investigation of age-related gene expression dynamics in the mouse corpus callosum, we compared RNA-seq data from 2 week-old and 12 week-old wild-type C57BL/6 J mice and identified the differentially expressed genes (e.g., Marcksl1, Chst3, C4b, Neat1, Ndrg1, Emid1, etc.) between these ages. Interestingly, we found that genes highly expressed in myelinating oligodendrocytes were upregulated in 12 week-old mice compared to 2 week-old mice, while genes highly expressed in oligodendrocyte precursor cells (OPCs) and newly formed oligodendrocytes were downregulated. Furthermore, by comparing these genes with the datasets from 20 week-old and 96 week-old mice, we identified novel sets of genes with age-dependent variations in the corpus callosum. These gene expression changes potentially affect key biological pathways and may be closely linked to age-related neurological disorders, including dementia and stroke. Therefore, our results provide an additional dataset to explore age-dependent gene expression dynamics of oligodendrocyte lineage cells in the corpus callosum.
Collapse
Affiliation(s)
- Tomonori Hoshino
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA.
| | - Hajime Takase
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Shintaro Kimura
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Norito Fukuda
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Josephine Lok
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Room 2401, Charlestown, MA, 02129-2000, USA.
| |
Collapse
|
31
|
Geppert J, Rohm M. Cancer cachexia: biomarkers and the influence of age. Mol Oncol 2024; 18:2070-2086. [PMID: 38414161 PMCID: PMC11467804 DOI: 10.1002/1878-0261.13590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 12/01/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024] Open
Abstract
Cancer cachexia (Ccx) is a complex metabolic condition characterized by pronounced muscle and fat wasting, systemic inflammation, weakness and fatigue. Up to 30% of cancer patients succumb directly to Ccx, yet therapies that effectively address this perturbed metabolic state are rare. In recent decades, several characteristics of Ccx have been established in mice and humans, of which we here highlight adipose tissue dysfunction, muscle wasting and systemic inflammation, as they are directly linked to biomarker discovery. To counteract cachexia pathogenesis as early as possible and mitigate its detrimental impact on anti-cancer treatments, identification and validation of clinically endorsed biomarkers assume paramount importance. Ageing was recently shown to affect both the validity of Ccx biomarkers and Ccx development, but the underlying mechanisms are still unknown. Thus, unravelling the intricate interplay between ageing and Ccx can help to counteract Ccx pathogenesis and tailor diagnostic and treatment strategies to individual needs.
Collapse
Affiliation(s)
- Julia Geppert
- Institute for Diabetes and CancerHelmholtz MunichNeuherbergGermany
- Joint Heidelberg‐IDC Translational Diabetes Program, Inner Medicine 1Heidelberg University HospitalGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Maria Rohm
- Institute for Diabetes and CancerHelmholtz MunichNeuherbergGermany
- Joint Heidelberg‐IDC Translational Diabetes Program, Inner Medicine 1Heidelberg University HospitalGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| |
Collapse
|
32
|
Niu K, Chang L, Zhang R, Jiang Y, Shen X, Lu X, Zhang S, Ma K, Zhao Z, Li M, Hou Y, Wu Y. Bazi Bushen mitigates age-related muscular atrophy by alleviating cellular senescence of skeletal muscle. J Tradit Complement Med 2024; 14:510-521. [PMID: 39262657 PMCID: PMC11385411 DOI: 10.1016/j.jtcme.2024.01.009] [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: 09/26/2023] [Revised: 01/06/2024] [Accepted: 01/21/2024] [Indexed: 09/13/2024] Open
Abstract
Background and aim Muscular atrophy is one of the most common age-related conditions characterized by the deterioration of skeletal muscle structures and impaired functions. It is associated with cellular senescence and chronic inflammation, which impair the function of muscle stem cells. Bazi Bushen (BZBS) is a patent compound Chinese medicine that has been shown to have anti-aging effects in various animal models. In this study, we investigated the effects and mechanisms of BZBS on muscular atrophy in naturally aged mice. Experimental procedure A muscular atrophy model of naturally aged mice (18 months) was employed with administration of BZBS (2 g/kg/d, 1 g/kg/d) and nicotinamide mononucleotide (NMN, 200 mg/kg/d). After six months of drug administration, muscle weight loss, muscle function and muscle histopathology were measured to evaluate the therapeutic effect of BZBS. The expression of cellular senescence, inflammatory and satellite cell-related factors were used to assess the effects of BZBS in inhibiting cellular senescence, reducing inflammation and improving muscle atrophy. Results and conclusion Compared with age matched natural aging mice, we found that BZBS improved muscle strength, mass, and morphology by reducing senescent cells, inflammatory cytokines, and intermyofiber fibrosis in aged muscle tissues. We also found that BZBS prevented the reduction of Pax7 positive stem cells and stimulated the activation and differentiation into myocytes. Our results suggest that BZBS might be a promising intervention in senile muscular atrophy.
Collapse
Affiliation(s)
- Kunxu Niu
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Liping Chang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- High-level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, 050035, China
| | - Runtao Zhang
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Yuning Jiang
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaogang Shen
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Xuan Lu
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Shixiong Zhang
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Kun Ma
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- High-level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, 050035, China
| | - Zhiqin Zhao
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Mengnan Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China
| | - Yunlong Hou
- Hebei Medical University, Shijiazhuang, 050017, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Yiling Wu
- Hebei Medical University, Shijiazhuang, 050017, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| |
Collapse
|
33
|
Hu L, Huang W, Liu B, Eklund EA. In Fanconi anemia, impaired accumulation of bone marrow neutrophils during emergency granulopoiesis induces hematopoietic stem cell stress. J Biol Chem 2024; 300:107548. [PMID: 38992437 PMCID: PMC11342097 DOI: 10.1016/j.jbc.2024.107548] [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: 02/05/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
Fanconi anemia (FA) is an inherited disorder of DNA repair due to mutation in one of 20+ interrelated genes that repair intrastrand DNA crosslinks and rescue collapsed or stalled replication forks. The most common hematologic abnormality in FA is anemia, but progression to bone marrow failure (BMF), clonal hematopoiesis, or acute myeloid leukemia may also occur. In prior studies, we found that Fanconi DNA repair is required for successful emergency granulopoiesis; the process for rapid neutrophil production during the innate immune response. Specifically, Fancc-/- mice did not develop neutrophilia in response to emergency granulopoiesis stimuli, but instead exhibited apoptosis of bone marrow hematopoietic stem cells and differentiating neutrophils. Repeated emergency granulopoiesis challenges induced BMF in most Fancc-/- mice, with acute myeloid leukemia in survivors. In contrast, we found equivalent neutrophilia during emergency granulopoiesis in Fancc-/-Tp53+/- mice and WT mice, without BMF. Since termination of emergency granulopoiesis is triggered by accumulation of bone marrow neutrophils, we hypothesize neutrophilia protects Fancc-/-Tp53+/- bone marrow from the stress of a sustained inflammation that is experienced by Fancc-/- mice. In the current work, we found that blocking neutrophil accumulation during emergency granulopoiesis led to BMF in Fancc-/-Tp53+/- mice, consistent with this hypothesis. Blocking neutrophilia during emergency granulopoiesis in Fancc-/-Tp53+/- mice (but not WT) impaired cell cycle checkpoint activity, also found in Fancc-/- mice. Mechanisms for loss of cell cycle checkpoints during infectious disease challenges may define molecular markers of FA progression, or suggest therapeutic targets for bone marrow protection in this disorder.
Collapse
Affiliation(s)
- Liping Hu
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Weiqi Huang
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, Illinois, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Bin Liu
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, Illinois, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Elizabeth A Eklund
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, Illinois, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown VA Medical Center, Chicago, Illinois, USA.
| |
Collapse
|
34
|
Nishiyama M, Kalambogias J, Imai F, Yang E, Lang S, de Nooij JC, Yoshida Y. Anatomical and functional analysis of the corticospinal tract in an FRDA mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601178. [PMID: 39005321 PMCID: PMC11244874 DOI: 10.1101/2024.06.28.601178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Friedreich's ataxia (FRDA) is one of the most common hereditary ataxias. It is caused by a GAA repeat in the first intron of the FXN gene, which encodes an essential mitochondrial protein. Patients suffer from progressive motor dysfunction due to the degeneration of mechanoreceptive and proprioceptive neurons in dorsal root ganglia (DRG) and cerebellar dentate nucleus neurons, especially at early disease stages. Postmortem analyses of FRDA patients also indicate pathological changes in motor cortex including in the projection neurons that give rise to the cortical spinal tract (CST). Yet, it remains poorly understood how early in the disease cortical spinal neurons (CSNs) show these alterations, or whether CSN/CST pathology resembles the abnormalities observed in other tissues affected by FXN loss. To address these questions, we examined CSN driven motor behaviors and pathology in the YG8JR FRDA mouse model. We find that FRDA mice show impaired motor skills, exhibit significant reductions in CSN functional output, and, among other pathological changes, show abnormal mitochondrial distributions in CSN neurons and CST axonal tracts. Moreover, some of these alterations were observed as early as two months of age, suggesting that CSN/CST pathology may be an earlier event in FRDA disease than previously appreciated. These studies warrant a detailed mechanistic understanding of how FXN loss impacts CSN health and functionality.
Collapse
Affiliation(s)
- Misa Nishiyama
- Burke Neurological Institute, White Plains, New York, United States
| | - John Kalambogias
- Burke Neurological Institute, White Plains, New York, United States
- Department of Neurology, Columbia University, New York, NY, USA
| | - Fumiyasu Imai
- Burke Neurological Institute, White Plains, New York, United States
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
| | - Emily Yang
- Burke Neurological Institute, White Plains, New York, United States
| | - Sonia Lang
- Burke Neurological Institute, White Plains, New York, United States
| | | | - Yutaka Yoshida
- Burke Neurological Institute, White Plains, New York, United States
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
35
|
Tarkhov AE, Lindstrom-Vautrin T, Zhang S, Ying K, Moqri M, Zhang B, Tyshkovskiy A, Levy O, Gladyshev VN. Nature of epigenetic aging from a single-cell perspective. NATURE AGING 2024; 4:854-870. [PMID: 38724733 DOI: 10.1038/s43587-024-00616-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 03/26/2024] [Indexed: 05/15/2024]
Abstract
Age-related changes in DNA methylation (DNAm) form the basis of the most robust predictors of age-epigenetic clocks-but a clear mechanistic understanding of exactly which aspects of aging are quantified by these clocks is lacking. Here, to clarify the nature of epigenetic aging, we juxtapose the dynamics of tissue and single-cell DNAm in mice. We compare these changes during early development with those observed during adult aging in mice, and corroborate our analyses with a single-cell RNA sequencing analysis within the same multiomics dataset. We show that epigenetic aging involves co-regulated changes as well as a major stochastic component, and this is consistent with transcriptional patterns. We further support the finding of stochastic epigenetic aging by direct tissue and single-cell DNAm analyses and modeling of aging DNAm trajectories with a stochastic process akin to radiocarbon decay. Finally, we describe a single-cell algorithm for the identification of co-regulated and stochastic CpG clusters showing consistent transcriptomic coordination patterns. Together, our analyses increase our understanding of the basis of epigenetic clocks and highlight potential opportunities for targeting aging and evaluating longevity interventions.
Collapse
Affiliation(s)
- Andrei E Tarkhov
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Retro Biosciences Inc., Redwood City, CA, USA.
| | - Thomas Lindstrom-Vautrin
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sirui Zhang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kejun Ying
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Obstetrics & Gynecology, Stanford School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Bohan Zhang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Orr Levy
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Wu X, Sun AR, Crawford R, Xiao Y, Wang Y, Prasadam I, Mao X. Inhibition of Leukotriene A 4 Hydrolase Suppressed Cartilage Degradation and Synovial Inflammation in a Mouse Model of Experimental Osteoarthritis. Cartilage 2024; 15:184-194. [PMID: 37086004 PMCID: PMC11368897 DOI: 10.1177/19476035231169940] [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: 12/18/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/23/2023] Open
Abstract
OBJECTIVE Chronic inflammation plays an important role in the osteoarthritis (OA) pathology but how this influence OA disease progression is unclear. Leukotriene B4 (LTB4) is a potent proinflammatory lipid mediator generated from arachidonic acid through the sequential activities of 5-lipoxygenase, 5-lipoxygenase-activating protein, Leukotriene A4 hydrolase (LTA4H) and its downstream product LTB4. The aim of this study is to investigate the involvement and the potential therapeutic target of the LTB4 pathway in OA disease progression. DESIGN Both clinical human cartilage samples (n = 7) and mice experimental OA models (n = 6) were used. The levels of LTA4H and leukotriene B4 receptor 1 were first examined using immunostaining in human OA/non-OA cartilage and mice experimental OA models. We also determined whether the LTA4H pathway was associated with cartilage degeneration and synovitis inflammation in OA mice models and human articular chondrocytes. RESULTS We found that both LTA4H and LTB4 receptor (BLT1) were highly expressed in human and mice OA cartilage. Inhibition of LTA4H suppressed cartilage degeneration and synovitis in OA mice model. Furthermore, inhibition of LTA4H promoted cartilage regeneration by upregulating chondrogenic genes expression such as aggrecan (ACAN), collagen 2A1 (COL2A1), and SRY-Box transcription factor 9 (SOX9). CONCLUSIONS Our results indicate that the LTA4H pathway is a crucial regulator of OA pathogenesis and suggest that LTA4H could be a therapeutic target in combat OA.
Collapse
Affiliation(s)
- Xiaoxin Wu
- Department of Orthopaedic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Centre for Biomedical Technologies, Faculty of Science and Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia RuJia Sun
- Centre for Biomedical Technologies, Faculty of Science and Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- School of Medicine and Dentistry, Griffith University, Brisbane, QLD, Australia
| | - Ross Crawford
- Centre for Biomedical Technologies, Faculty of Science and Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Orthopaedic Department, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Yin Xiao
- School of Medicine and Dentistry, Griffith University, Brisbane, QLD, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yanping Wang
- Health Management Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Indira Prasadam
- Centre for Biomedical Technologies, Faculty of Science and Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Xinzhan Mao
- Department of Orthopaedic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
37
|
Aly R, Darwish S, Bala N, Ebrahim A, Shoemaker LR, McCray J, Garrett TJ, Alli AA. Functional and metabolomic analysis of urinary extracellular vesicles from juvenile mice with renal compensatory hypertrophy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167096. [PMID: 38499276 PMCID: PMC11632668 DOI: 10.1016/j.bbadis.2024.167096] [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: 11/22/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024]
Abstract
Unilateral nephrectomy, a procedure reducing kidney mass, triggers a compensatory response in the remaining kidney, increasing its size and function to maintain a normal glomerular filtration rate (GFR). Recent research has highlighted the role of extracellular vesicles (EVs) in renal physiology and disease, although their involvement in unilateral nephrectomy has been underexplored. In this study, unilateral nephrectomy was performed on young mice, and urinary extracellular vesicles (uEVs) characterization and cargo were analyzed. Kidney volume increased significantly post-nephrectomy, demonstrating compensatory hypertrophy. Serum creatinine, cystatin C, and urinary electrolytes concentrations were similar in both nephrectomized and control groups. Western blot analysis revealed upregulation of sodium-glucose cotransporter 2 (SGLT2) and sodium chloride cotransporter (NCC), and downregulation of sodium‑potassium-chloride co-transporter (NKCC2) and epithelial sodium channel (ENaC) in the nephrectomized group. Metabolomic analysis of uEVs showed an enrichment of certain metabolites, including citrate and stachydrine. Interestingly, uEVs from the nephrectomized group demonstrated a protective effect, downregulating signal transducer and activator of transcription 3 (STAT3) and reducing reactive oxygen species (ROS) in renal proximal cells, compared to uEVs from the control group. This study suggests that uEVs contain bioactive components capable of inducing protective, anti-inflammatory, anti-fibrinolytic, and antioxidative effects in renal cells. These findings contribute to our understanding of uEVs' role in renal compensatory mechanisms after unilateral nephrectomy and may hold promise for future therapeutic interventions in renal diseases.
Collapse
Affiliation(s)
- Rasha Aly
- Departement of Pediatrics, Division of Pediatric Nephrology, University of Florida, 32610, United States of America
| | - Sara Darwish
- Department of Physiology and Aging, College of Medicine, University of Florida, 32610, United States of America
| | - Niharika Bala
- Department of Physiology and Aging, College of Medicine, University of Florida, 32610, United States of America
| | - Areej Ebrahim
- Department of Physiology and Aging, College of Medicine, University of Florida, 32610, United States of America
| | - Lawrence R Shoemaker
- Departement of Pediatrics, Division of Pediatric Nephrology, University of Florida, 32610, United States of America
| | - Joel McCray
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610, United States of America
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610, United States of America
| | - Abdel A Alli
- Department of Physiology and Aging, College of Medicine, University of Florida, 32610, United States of America; Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, 32610, United States of America.
| |
Collapse
|
38
|
Ruiz-Lara G, Costa-Silva TA, Muso-Cachumba JJ, Cevallos Espinel J, Fontes MG, Garcia-Maya M, Rahman KM, Rangel-Yagui CDO, Monteiro G. Nonclinical Evaluation of Single-Mutant E. coli Asparaginases Obtained by Double-Mutant Deconvolution: Improving Toxicological, Immune and Inflammatory Responses. Int J Mol Sci 2024; 25:6008. [PMID: 38892196 PMCID: PMC11172649 DOI: 10.3390/ijms25116008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Acute lymphoblastic leukaemia is currently treated with bacterial L-asparaginase; however, its side effects raise the need for the development of improved and efficient novel enzymes. Previously, we obtained low anti-asparaginase antibody production and high serum enzyme half-life in mice treated with the P40S/S206C mutant; however, its specific activity was significantly reduced. Thus, our aim was to test single mutants, S206C and P40S, through in vitro and in vivo assays. Our results showed that the drop in specific activity was caused by P40S substitution. In addition, our single mutants were highly stable in biological environment simulation, unlike the double-mutant P40S/S206C. The in vitro cell viability assay demonstrated that mutant enzymes have a higher cytotoxic effect than WT on T-cell-derived ALL and on some solid cancer cell lines. The in vivo assays were performed in mice to identify toxicological effects, to evoke immunological responses and to study the enzymes' pharmacokinetics. From these tests, none of the enzymes was toxic; however, S206C elicited lower physiological changes and immune/allergenic responses. In relation to the pharmacokinetic profile, S206C exhibited twofold higher activity than WT and P40S two hours after injection. In conclusion, we present bioengineered E. coli asparaginases with high specific enzyme activity and fewer side effects.
Collapse
Affiliation(s)
- Grace Ruiz-Lara
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (G.R.-L.); (J.J.M.-C.); (M.G.F.); (C.d.O.R.-Y.)
| | - Tales A. Costa-Silva
- Center for Natural and Human Sciences, Federal University of ABC, Santo André 09210-580, SP, Brazil;
| | - Jorge Javier Muso-Cachumba
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (G.R.-L.); (J.J.M.-C.); (M.G.F.); (C.d.O.R.-Y.)
| | | | - Marina Gabriel Fontes
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (G.R.-L.); (J.J.M.-C.); (M.G.F.); (C.d.O.R.-Y.)
| | - Mitla Garcia-Maya
- Randall Division of Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK;
| | | | - Carlota de Oliveira Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (G.R.-L.); (J.J.M.-C.); (M.G.F.); (C.d.O.R.-Y.)
| | - Gisele Monteiro
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (G.R.-L.); (J.J.M.-C.); (M.G.F.); (C.d.O.R.-Y.)
| |
Collapse
|
39
|
Ramnauth AD, Tippani M, Divecha HR, Papariello AR, Miller RA, Nelson ED, Pattie EA, Kleinman JE, Maynard KR, Collado-Torres L, Hyde TM, Martinowich K, Hicks SC, Page SC. Spatiotemporal analysis of gene expression in the human dentate gyrus reveals age-associated changes in cellular maturation and neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.20.567883. [PMID: 38045413 PMCID: PMC10690172 DOI: 10.1101/2023.11.20.567883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The dentate gyrus of the hippocampus is important for many cognitive functions, including learning, memory, and mood. Here, we investigated age-associated changes in transcriptome-wide spatial gene expression in the human dentate gyrus across the lifespan. Genes associated with neurogenesis and the extracellular matrix were enriched in infants, while gene markers of inhibitory neurons and cell proliferation showed increases and decreases in post-infancy, respectively. While we did not find evidence for neural proliferation post-infancy, we did identify molecular signatures supporting protracted maturation of granule cells. We also identified a wide-spread hippocampal aging signature and an age-associated increase in genes related to neuroinflammation. Our findings suggest major changes to the putative neurogenic niche after infancy and identify molecular foci of brain aging in glial and neuropil enriched tissue.
Collapse
|
40
|
Hinks A, Dalton BE, Mashouri P, Flewwelling LD, Pyle WG, Cheng AJ, Power GA. Time course changes in in vivo muscle mechanical function and Ca 2+ regulation of force following experimentally induced gradual ovarian failure in mice. Exp Physiol 2024; 109:711-728. [PMID: 38500268 PMCID: PMC11061627 DOI: 10.1113/ep091735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/05/2024] [Indexed: 03/20/2024]
Abstract
The abrupt cessation of ovarian hormone release is associated with declines in muscle contractile function, yet the impact of gradual ovarian failure on muscle contractility across peri-, early- and late-stage menopause remains unclear. In this study, a 4-vinylcyclohexene diepoxide (VCD)-induced ovarian failure mouse model was used to examine time course changes in muscle mechanical function. Plantar flexors of female mice (VCD: n = 10; CON: n = 8) were assessed at 40 (early perimenopause), 80 (late perimenopause), 120 (menopause onset) and 176 (late menopause) days post-initial VCD injection. A torque-frequency relationship was established across a range of frequencies (10-200 Hz). Isotonic dynamic contractions were elicited against relative loads (10-80% maximal isometric torque) to determine the torque-velocity-power relationship. Mice then performed a fatigue task using intermittent 100 Hz isometric contractions until torque dropped by 60%. Recovery of twitch, 10 Hz and 100 Hz torque were tracked for 10 min post-task failure. Additionally, intact muscle fibres from the flexor digitorum brevis underwent a fatigue task (50 repetitions at 70 Hz), and 10 and 100 Hz tetanic [Ca2+] were monitored for 10 min afterward. VCD mice exhibited 16% lower twitch torque than controls across all time points. Apart from twitch torque, 10 Hz torque and 10 Hz tetanic [Ca2+], where VCD showed greater values relative to pre-fatigue during recovery, no significant differences were observed between control and VCD mice during recovery. These results indicate that gradual ovarian failure has minimal detriments to in vivo muscle mechanical function, with minor alterations observed primarily for low-frequency stimulation during recovery from fatigue.
Collapse
Affiliation(s)
- Avery Hinks
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| | - Benjamin E. Dalton
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| | - Parastoo Mashouri
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| | - Luke D. Flewwelling
- Muscle Health Research Centre, School of Kinesiology and Health Sciences, Faculty of HealthYork UniversityTorontoCanada
| | - William Glen Pyle
- IMPART Team Canada, Dalhousie MedicineDalhousie UniversitySaint JohnNew BrunswickCanada
| | - Arthur J. Cheng
- Muscle Health Research Centre, School of Kinesiology and Health Sciences, Faculty of HealthYork UniversityTorontoCanada
| | - Geoffrey A. Power
- Department of Human Health and Nutritional Sciences, College of Biological SciencesUniversity of GuelphGuelphOntarioCanada
| |
Collapse
|
41
|
Ma K, Li M, Song Y, Mao X, Zhang S, Zhang Y, Jiang Y, Ji C, Niu K, Li H, Hou Y, Wei C. Bazi Bushen alleviates reproductive aging in aged male mice. Chin J Nat Med 2024; 22:416-425. [PMID: 38796215 DOI: 10.1016/s1875-5364(24)60639-4] [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: 06/19/2023] [Indexed: 05/28/2024]
Abstract
Bazi Bushen (BZBS), a traditional Chinese medicine (TCM), has demonstrated therapeutic efficacy in testicular dysfunction within D-galactose and NaNO2 mouse models. This study aimed to ascertain if BZBS could also mitigate the decline in testicular function associated with natural aging. Therefore, male aged mice were employed to evaluate the preventive effects of BZBS on male reproductive aging. This was achieved by assessing sex hormone production, testicular histomorphology, and spermatogenesis. Relative to the untreated aged control group, BZBS administration elevated the levels of sex hormones and spermatocyte populations and preserved normal testicular structure in aged mice. Notably, spermatogenesis was maintained. Further analyses, including malondialdehyde (MDA) assays and real-time PCR, indicated that BZBS diminished testicular oxidative stress and the inflammatory burden. Corroborating these findings, mice treated with BZBS exhibited reductions in the populations of senescent and apoptotic cells within the seminiferous tubules, suggesting alleviated cellular damage. In contrast, we observed that rapamycin, a drug known for its longevity benefits, induced excessive testicular apoptosis and did not decrease lipid peroxidation. Collectively, our results highlight BZBS's promising clinical potential in counteracting male reproductive aging, underlining its mechanisms of action.
Collapse
Affiliation(s)
- Kun Ma
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Mengnan Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China; Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang 050035, China
| | - Yahui Song
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China
| | - Xinjing Mao
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Hebei Provincial Key Laboratory of Luobing, Shijiazhuang 050035, China
| | - Shaolan Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; Key Disciplines of State Administration of TCM for Luobing, Shijiazhuang 050035, China
| | - Yaping Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; Key Disciplines of State Administration of TCM for Luobing, Shijiazhuang 050035, China
| | - Yuning Jiang
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Hebei Provincial Key Laboratory of Luobing, Shijiazhuang 050035, China
| | - Chuanyuan Ji
- College of Traditional Chinese Medicine·College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Hebei Provincial Key Laboratory of Luobing, Shijiazhuang 050035, China
| | - Kunxu Niu
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang 050035, China; Hebei Provincial Key Laboratory of Luobing, Shijiazhuang 050035, China
| | - Hongrong Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China; Hebei Provincial Key Laboratory of Luobing, Shijiazhuang 050035, China
| | - Yunlong Hou
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China; Hebei Provincial Key Laboratory of Luobing, Shijiazhuang 050035, China.
| | - Cong Wei
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China.
| |
Collapse
|
42
|
Fabritz L, Fortmueller L, Gehmlich K, Kant S, Kemper M, Kucerova D, Syeda F, Faber C, Leube RE, Kirchhof P, Krusche CA. Endurance Training Provokes Arrhythmogenic Right Ventricular Cardiomyopathy Phenotype in Heterozygous Desmoglein-2 Mutants: Alleviation by Preload Reduction. Biomedicines 2024; 12:985. [PMID: 38790949 PMCID: PMC11117820 DOI: 10.3390/biomedicines12050985] [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: 11/06/2023] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Desmoglein-2 mutations are detected in 5-10% of patients with arrhythmogenic right ventricular cardiomyopathy (ARVC). Endurance training accelerates the development of the ARVC phenotype, leading to earlier arrhythmic events. Homozygous Dsg2 mutant mice develop a severe ARVC-like phenotype. The phenotype of heterozygous mutant (Dsg2mt/wt) or haploinsufficient (Dsg20/wt) mice is still not well understood. To assess the effects of age and endurance swim training, we studied cardiac morphology and function in sedentary one-year-old Dsg2mt/wt and Dsg20/wt mice and in young Dsg2mt/wt mice exposed to endurance swim training. Cardiac structure was only occasionally affected in aged Dsg20/wt and Dsg2mt/wt mice manifesting as small fibrotic foci and displacement of Connexin 43. Endurance swim training increased the right ventricular (RV) diameter and decreased RV function in Dsg2mt/wt mice but not in wild types. Dsg2mt/wt hearts showed increased ventricular activation times and pacing-induced ventricular arrhythmia without obvious fibrosis or inflammation. Preload-reducing therapy during training prevented RV enlargement and alleviated the electrophysiological phenotype. Taken together, endurance swim training induced features of ARVC in young adult Dsg2mt/wt mice. Prolonged ventricular activation times in the hearts of trained Dsg2mt/wt mice are therefore a potential mechanism for increased arrhythmia risk. Preload-reducing therapy prevented training-induced ARVC phenotype pointing to beneficial treatment options in human patients.
Collapse
Affiliation(s)
- Larissa Fabritz
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center, University Hospital Hamburg Eppendorf, 20246 Hamburg, Germany; (L.F.); (P.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Lisa Fortmueller
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center, University Hospital Hamburg Eppendorf, 20246 Hamburg, Germany; (L.F.); (P.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX1 2JD, UK
| | - Sebastian Kant
- Institute for Molecular and Cellular Anatomy (MOCA), RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (R.E.L.)
| | - Marcel Kemper
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Dana Kucerova
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Fahima Syeda
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
| | - Cornelius Faber
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Muenster, 48149 Münster, Germany;
| | - Rudolf E. Leube
- Institute for Molecular and Cellular Anatomy (MOCA), RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (R.E.L.)
| | - Paulus Kirchhof
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center, University Hospital Hamburg Eppendorf, 20246 Hamburg, Germany; (L.F.); (P.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
| | - Claudia A. Krusche
- Institute for Molecular and Cellular Anatomy (MOCA), RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (R.E.L.)
| |
Collapse
|
43
|
Yang S, Liu P, Zhang Y, Xu H, Lan J, Jiang H, Jin G, Bai X. Single-cell transcriptome atlas in C57BL/6 mice encodes morphological phenotypes in the aging kidneys. BMC Nephrol 2024; 25:137. [PMID: 38641839 PMCID: PMC11031943 DOI: 10.1186/s12882-024-03514-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/20/2024] [Indexed: 04/21/2024] Open
Abstract
C57BL/6 mice are frequently utilized as murine models with the desired genetic background for altertion in multiple research contexts. So far, there is still a lack of comprehensive kidney morphology and single-cell transcriptome atlas at all stages of growth of C57BL/6 mice. To provide an interactive set of reference standards for the scientific community, we performed the current study to investigate the kidney's development throughout the capillary-loop stage until senescence. Eight groups, with five to six mice each, represented embryonic stage (embryos 18.5 days), suckling period (1 day after birth), juvenile stage (1 month old), adulthood (containing 3 months old, 6 months old and 10 months old), reproductive senescence stage (20 months old), and post-senescence stage (30 months old), respectively. With age, the thickness of the glomerular basement membrane (GBM) was increased. Notably, GBM knobs appeared at three months and became frequent with age. Using single-cell transcriptome data, we evaluated how various biological process appear in particular cell types and investigated the potential mechanism of formation of GBM konbs. In conclusion, having access to detailed kidney morphology and single-cell transcriptome maps from C57BL/6 mice at various developmental stages of C57BL/6 mice would be a novel and major resource for biological research and testing of prospective therapeutic approaches.
Collapse
Affiliation(s)
- Shanzhi Yang
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China
| | - Peimin Liu
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China
| | - Yan Zhang
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China
| | - Haosen Xu
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China
| | - Jinyi Lan
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China
| | - Huan Jiang
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China
| | - Guoxiang Jin
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China.
| | - Xiaoyan Bai
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, China.
| |
Collapse
|
44
|
Tang J, Zhang J, Zhang G, Peng W, Ling N, Zhou Y, Xu H, Ren H, Chen M. Stat3 activation-triggered transcriptional networks govern the early stage of HBV-induced hepatic inflammation. mBio 2024; 15:e0306823. [PMID: 38440978 PMCID: PMC11005361 DOI: 10.1128/mbio.03068-23] [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: 11/17/2023] [Accepted: 02/01/2024] [Indexed: 03/06/2024] Open
Abstract
The chronic carrier state of the hepatitis B virus (HBV) often leads to the development of liver inflammation as carriers age. However, the exact mechanisms that trigger this hepatic inflammation remain poorly defined. We analyzed the sequential processes during the onset of liver inflammation based on time-course transcriptome and transcriptional regulatory networks in an HBV transgenic (HBV-Tg) mice model and chronic HBV-infected (CHB) patients (data from GSE83148). The key transcriptional factor (TF) responsible for hepatic inflammation occurrence was identified and then validated both in HBV-Tg mice and liver specimens from young CHB patients. By time-course analysis, an early stage of hepatic inflammation was demonstrated in 3-month-old HBV-Tg mice: a marked upregulation of genes related to inflammation (Saa1/2, S100a8/9/11, or Il1β), innate immunity (Tlr2, Tlr7, or Tlr8), and cells chemotaxis (Ccr2, Cxcl1, Cxcl13, or Cxcl14). Within CHB samples, a unique early stage of inflammation activation was discriminated from immune tolerance and immune activation groups based on distinct gene expression patterns. Enhanced activation of TF Stat3 was strongly associated with increased inflammatory gene expression in this early stage of inflammation. Expression of phosphorylated Stat3 was higher in liver specimens from young CHB patients with relatively higher alanine aminotransferase levels. Specific inhibition of Stat3 activation significantly attenuated the degree of liver inflammation, the expression of inflammation-related genes, and the inflammatory monocytes and macrophages in 3-month-old HBV-Tg mice. Stat3 activation is essential for hepatic inflammation occurrence and is a novel indicator of early-stage immune activation in chronic HBV carriers. IMPORTANCE Until now, it remains a mystery that chronic hepatitis B virus (HBV)-infected patients in the "immune tolerance phase" will transition to the "immune activation phase" as they age. In this study, we reveal that Stat3 activation-triggered hepatic transcriptional alterations are distinctive characteristics of the early stage of immune/inflammation activation in chronic HBV infection. For the first time, we discover a mechanism that might trigger the transition from immune tolerance to immune activation in chronic HBV carriers.
Collapse
Affiliation(s)
- Jinglin Tang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Transfusion Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jiaxuan Zhang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Laboratory Medicine, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Gaoli Zhang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenhui Peng
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ning Ling
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingzhi Zhou
- Department of Infection, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmei Xu
- Department of Infection, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Ren
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Chen
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
45
|
Lloyd EG, Henríquez JA, Biffi G. Modelling the micro- and macro- environment of pancreatic cancer: from patients to pre-clinical models and back. Dis Model Mech 2024; 17:dmm050624. [PMID: 38639944 PMCID: PMC11051978 DOI: 10.1242/dmm.050624] [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: 04/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with very low survival rates. Over the past 50 years, improvements in PDAC survival have significantly lagged behind the progress made in other cancers. PDAC's dismal prognosis is due to typical late-stage diagnosis combined with lack of effective treatments and complex mechanisms of disease. We propose that improvements in survival are partly hindered by the current focus on largely modelling and targeting PDAC as one disease, despite it being heterogeneous. Implementing new disease-representative pre-clinical mouse models that capture this complexity could enable the development of transformative therapies. Specifically, these models should recapitulate human PDAC late-stage biology, heterogeneous genetics, extensive non-malignant stroma, and associated risk factors and comorbidities. In this Perspective, we focus on how pre-clinical mouse models could be improved to exemplify key features of PDAC micro- and macro- environments, which would drive clinically relevant patient stratification, tailored treatments and improved survival.
Collapse
Affiliation(s)
- Eloise G. Lloyd
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Joaquín Araos Henríquez
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
46
|
Xie P, Li P, Zhu X, Chen D, Ommati MM, Wang H, Han L, Xu S, Sun P. Hepatotoxic of polystyrene microplastics in aged mice: Focus on the role of gastrointestinal transformation and AMPK/FoxO pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 917:170471. [PMID: 38296072 DOI: 10.1016/j.scitotenv.2024.170471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/05/2024]
Abstract
Microplastic (MP) toxicity has attracted widespread attention, whereas before triggering hepatotoxicity, ingested MPs first undergo transportation and digestion processes in the gastrointestinal tract, possibly interacting with the gastrointestinal contents (GIC). More alarming is the need for more understanding of how this process may impact the liver health of aged animals. This study selected old mice. Firstly, we incubated polystyrene microplastics (PS-MPs, 1 μm) with GIC extract. The results of SEM/EDS indicated a structural alteration in PS-MPs. Additionally, impurities resembling corona, rich in heteroatoms (O, N, and S), were observed. This resulted in an enhanced aggregating phenomenon of MPs. We conducted a 10-day experiment exposing aged mice to four concentrations of PS-MPs, ranging from 1 × 103 to 1 × 1012 particles/L. Subsequent measurements of tissue pathology and body and organ weights were conducted, revealing alterations in liver structure. In the liver, 12 crucial metabolites were found by LC-MS technology, including purines, lipids, and amino acids. The AMPK/FoxO pathway was enriched, activated, and validated in western blotting results. We also comprehensively examined the innate immune system, inflammatory factors, and oxidative stress indicators. The results indicated decreased C3 levels, stable C4 levels, inflammatory factors (IL-6 and IL-8), and antioxidant enzymes were increased to varying degrees. PS-MPs also caused DNA oxidative damage. These toxic effects exhibited a specific dose dependence. Overall, after the formation of the gastrointestinal corona, PS-MPs subsequently impact various cellular processes, such as cycle arrest (p21), leading to hepatic and health crises in the elderly. The presence of gastrointestinal coronas also underscores the MPs' morphology and characteristics, which should be distinguished after ingestion.
Collapse
Affiliation(s)
- Pengfei Xie
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Pengcheng Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Xiaoshan Zhu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Deshan Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Mohammad Mehdi Ommati
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Hongwei Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Lei Han
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Shixiao Xu
- Northwest Institute of Plateau Biology Chinese Academy of Sciences, Xining, Qinghai 810008, China
| | - Ping Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China.
| |
Collapse
|
47
|
Surico PL, Narimatsu A, Forouzanfar K, Singh RB, Shoushtari S, Dana R, Blanco T. Effects of Diabetes Mellitus on Corneal Immune Cell Activation and the Development of Keratopathy. Cells 2024; 13:532. [PMID: 38534376 PMCID: PMC10969384 DOI: 10.3390/cells13060532] [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: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
Diabetes mellitus (DM) is one of the most prevalent diseases globally, and its prevalence is rapidly increasing. Most patients with a long-term history of DM present with some degree of keratopathy (DK). Despite its high incidence, the underlying inflammatory mechanism of DK has not been elucidated yet. For further insights into the underlying immunopathologic processes, we utilized streptozotocin-induced mice to model type 1 DM (T1D) and B6.Cg-Lepob/J mice to model type 2 DM (T2D). We evaluated the animals for the development of clinical manifestations of DK. Four weeks post-induction, the total frequencies of corneal CD45+CD11b+Ly-6G- myeloid cells, with enhanced gene and protein expression levels for the proinflammatory cytokines TNF-α and IL-1β, were higher in both T1D and T2D animals. Additionally, the frequencies of myeloid cells/mm2 in the sub-basal neural plexus (SBNP) were significantly higher in T1D and T2D compared to non-diabetic mice. DK clinical manifestations were observed four weeks post-induction, including significantly lower tear production, corneal sensitivity, and epitheliopathy. Nerve density in the SBNP and intraepithelial terminal endings per 40x field were lower in both models compared to the normal controls. The findings of this study indicate that DM alters the immune quiescent state of the cornea during disease onset, which may be associated with the progressive development of the clinical manifestations of DK.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tomas Blanco
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; (P.L.S.); (A.N.); (K.F.); (R.B.S.); (S.S.); (R.D.)
| |
Collapse
|
48
|
Liu Y, Xiao J, Cai J, Li R, Sui X, Zhang J, Lu T, Chen H, Chen G, Li H, Jiang C, Zhao X, Xiao C, Lei Y, Yao J, Lv G, Liang J, Zhang Y, Yang JR, Zheng J, Yang Y. Single-cell immune profiling of mouse liver aging reveals Cxcl2+ macrophages recruit neutrophils to aggravate liver injury. Hepatology 2024; 79:589-605. [PMID: 37695548 PMCID: PMC10871588 DOI: 10.1097/hep.0000000000000590] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/21/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND AND AIMS Immune cells play a crucial role in liver aging. However, the impact of dynamic changes in the local immune microenvironment on age-related liver injury remains poorly understood. We aimed to characterize intrahepatic immune cells at different ages to investigate key mechanisms associated with liver aging. APPROACH AND RESULTS We carried out single-cell RNA sequencing on mouse liver tissues at 4 different ages, namely, the newborn, suckling, young, and aged stages. The transcriptomic landscape, cellular classification, and intercellular communication were analyzed. We confirmed the findings by multiplex immunofluorescence staining, flow cytometry, in vitro functional experiments, and chimeric animal models. Nine subsets of 89,542 immune cells with unique properties were identified, of which Cxcl2+ macrophages within the monocyte/macrophage subset were preferentially enriched in the aged liver. Cxcl2+ macrophages presented a senescence-associated secretory phenotype and recruited neutrophils to the aged liver through the CXCL2-CXCR2 axis. Through the secretion of IL-1β and TNF-α, Cxcl2+ macrophages stimulated neutrophil extracellular traps formation. Targeting the CXCL2-CXCR2 axis limited the neutrophils migration toward the liver and attenuated age-related liver injury. Moreover, the relationship between Cxcl2+ macrophages and neutrophils in age-related liver injury was further validated by human liver transplantation samples. CONCLUSIONS This in-depth study illustrates that the mechanism of Cxcl2+ macrophage-driven neutrophil activation involves the CXCL2-CXCR2 axis and provides a potential therapeutic strategy for age-related liver injury.
Collapse
Affiliation(s)
- Yasong Liu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jiaqi Xiao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jianye Cai
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Rong Li
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Xin Sui
- Surgical ICU, The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jiebin Zhang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Tongyu Lu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Haitian Chen
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Guihua Chen
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Haibo Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Chenhao Jiang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Xuegang Zhao
- Surgical ICU, The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Cuicui Xiao
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University; Guangzhou, China
| | - Yunguo Lei
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jia Yao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Guo Lv
- Biological Treatment Center, The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jinliang Liang
- Organ Transplantation Research Center of Guangdong Province Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yingcai Zhang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Jian-Rong Yang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-sen University; Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University; Guangzhou, China
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine; Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, the Third Affiliated Hospital of Sun Yat-sen University; Guangzhou, China
| |
Collapse
|
49
|
Özalay Ö, Mediavilla T, Giacobbo BL, Pedersen R, Marcellino D, Orädd G, Rieckmann A, Sultan F. Longitudinal monitoring of the mouse brain reveals heterogenous network trajectories during aging. Commun Biol 2024; 7:210. [PMID: 38378942 PMCID: PMC10879497 DOI: 10.1038/s42003-024-05873-8] [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: 05/30/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024] Open
Abstract
The human aging brain is characterized by changes in network efficiency that are currently best captured through longitudinal resting-state functional MRI (rs-fMRI). These studies however are challenging due to the long human lifespan. Here we show that the mouse animal model with a much shorter lifespan allows us to follow the functional network organization over most of the animal's adult lifetime. We used a longitudinal study of the functional connectivity of different brain regions with rs-fMRI under anesthesia. Our analysis uncovers network modules similar to those reported in younger mice and in humans (i.e., prefrontal/default mode network (DMN), somatomotor and somatosensory networks). Statistical analysis reveals different patterns of network reorganization during aging. Female mice showed a pattern akin to human aging, with de-differentiation of the connectome, mainly due to increases in connectivity of the prefrontal/DMN cortical networks to other modules. Our male cohorts revealed heterogenous aging patterns with only one group confirming the de- differentiation, while the majority showed an increase in connectivity of the somatomotor cortex to the Nucleus accumbens. In summary, in line with human work, our analysis in mice supports the concept of de-differentiation in the aging mammalian brain and reveals additional trajectories in aging mice networks.
Collapse
Affiliation(s)
- Özgün Özalay
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden
| | - Tomas Mediavilla
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden
| | - Bruno Lima Giacobbo
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Robin Pedersen
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden
| | - Daniel Marcellino
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden
| | - Greger Orädd
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden
| | - Anna Rieckmann
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden
- Department of Diagnostics and Intervention, Radiation Physics, Umeå University, 90 187, Umeå, Sweden
- Institute for Psychology, University of the Bundeswehr Munich, Neubiberg, Germany
| | - Fahad Sultan
- Department of Medical and Translational Biology, Umeå University, 90 187, Umeå, Sweden.
| |
Collapse
|
50
|
Mishra DK, Popovski D, Morris SM, Bondoc A, Senthil Kumar S, Girard EJ, Rutka J, Fouladi M, Huang A, Olson JM, Drissi R. Preclinical pediatric brain tumor models for immunotherapy: Hurdles and a way forward. Neuro Oncol 2024; 26:226-235. [PMID: 37713135 PMCID: PMC10836771 DOI: 10.1093/neuonc/noad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Indexed: 09/16/2023] Open
Abstract
Brain tumors are the most common solid tumor in children and the leading cause of cancer-related deaths. Over the last few years, improvements have been made in the diagnosis and treatment of children with Central Nervous System tumors. Unfortunately, for many patients with high-grade tumors, the overall prognosis remains poor. Lower survival rates are partly attributed to the lack of efficacious therapies. The advent and success of immune checkpoint inhibitors (ICIs) in adults have sparked interest in investigating the utility of these therapies alone or in combination with other drug treatments in pediatric patients. However, to achieve improved clinical outcomes, the establishment and selection of relevant and robust preclinical pediatric high-grade brain tumor models is imperative. Here, we review the information that influenced our model selection as we embarked on an international collaborative study to test ICIs in combination with epigenetic modifying agents to enhance adaptive immunity to treat pediatric brain tumors. We also share challenges that we faced and potential solutions.
Collapse
Affiliation(s)
- Deepak Kumar Mishra
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Dean Popovski
- Arthur and Sonia Labatt Brain Tumor Research Centre, Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Onatario, Canada
| | - Shelli M Morris
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Andrew Bondoc
- Arthur and Sonia Labatt Brain Tumor Research Centre, Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Onatario, Canada
| | - Shiva Senthil Kumar
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Emily J Girard
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - James Rutka
- Division of Neurosurgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maryam Fouladi
- Pediatric Neuro-Oncology Program, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Annie Huang
- Arthur and Sonia Labatt Brain Tumor Research Centre, Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Onatario, Canada
| | - James M Olson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Rachid Drissi
- Center for Childhood Cancer Research, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|