151
|
Sheng L, Shields EJ, Gospocic J, Sorida M, Ju L, Byrns CN, Carranza F, Berger SL, Bonini N, Bonasio R. Ensheathing glia promote increased lifespan and healthy brain aging. Aging Cell 2023; 22:e13803. [PMID: 36840361 PMCID: PMC10186613 DOI: 10.1111/acel.13803] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/26/2023] Open
Abstract
Glia have an emergent role in brain aging and disease. In the Drosophila melanogaster brain, ensheathing glia function as phagocytic cells and respond to acute neuronal damage, analogous to mammalian microglia. We previously reported changes in glia composition over the life of ants and fruit flies, including a decline in the relative proportion of ensheathing glia with time. How these changes influence brain health and life expectancy is unknown. Here, we show that ensheathing glia but not astrocytes decrease in number during Drosophila melanogaster brain aging. The remaining ensheathing glia display dysregulated expression of genes involved in lipid metabolism and apoptosis, which may lead to lipid droplet accumulation, cellular dysfunction, and death. Inhibition of apoptosis rescued the decline of ensheathing glia with age, improved the neuromotor performance of aged flies, and extended lifespan. Furthermore, an expanded ensheathing glia population prevented amyloid-beta accumulation in a fly model of Alzheimer's disease and delayed the premature death of the diseased animals. These findings suggest that ensheathing glia play a vital role in regulating brain health and animal longevity.
Collapse
Affiliation(s)
- Lihong Sheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Emily J. Shields
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Urology and Institute of NeuropathologyMedical Center–University of FreiburgFreiburgGermany
| | - Janko Gospocic
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Urology and Institute of NeuropathologyMedical Center–University of FreiburgFreiburgGermany
| | - Masato Sorida
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Linyang Ju
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - China N. Byrns
- Medical Scientist Training ProgramUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Neuroscience Graduate GroupUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Faith Carranza
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Shelley L. Berger
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of GeneticsUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Nancy Bonini
- Neuroscience Graduate GroupUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of BiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Roberto Bonasio
- Epigenetics InstituteUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Department of Urology and Institute of NeuropathologyMedical Center–University of FreiburgFreiburgGermany
| |
Collapse
|
152
|
Lushington R, Camilli S, Pascual F, Lockey RF, Kolliputi N. EP2 inhibition restores myeloid metabolism and reverses cognitive decline. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100082. [PMID: 37780795 PMCID: PMC10509962 DOI: 10.1016/j.jacig.2023.100082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 10/03/2023]
Abstract
Nonsteroidal anti-inflammatory drugs alleviate pain and inflammation by inhibiting the cyclooxygenase pathway. This pathway has various downstream effects, some of which are beneficial. Prostaglandin E2 is a key downstream product in the cyclooxygenase pathway that modulates inflammation. A correlation between aging and increased expression of the prostaglandin E2 receptor, EP2, has been associated with inflammatory processes, cognitive aging, angiogenesis, and tumorigenesis. Therefore, inhibition of EP2 could lead to therapeutic effects and be more selective than inhibiting cyclooxygenase-2. Studies suggest that inhibition of EP2 restores age-associated spatial memory deficits and synaptic proteins and impairs tumorigenesis. The data indicate that EP2 signaling is important in myeloid cell metabolism and support its candidacy as a therapeutic target.
Collapse
Affiliation(s)
- Ryan Lushington
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa
| | - Samuel Camilli
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa
| | - Francisco Pascual
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa
| | - Richard F. Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
153
|
Abstract
Over the last decade, immunometabolism has emerged as a novel interdisciplinary field of research and yielded significant fundamental insights into the regulation of immune responses. Multiple classical approaches to interrogate immunometabolism, including bulk metabolic profiling and analysis of metabolic regulator expression, paved the way to appreciating the physiological complexity of immunometabolic regulation in vivo. Studying immunometabolism at the systems level raised the need to transition towards the next-generation technology for metabolic profiling and analysis. Spatially resolved metabolic imaging and computational algorithms for multi-modal data integration are new approaches to connecting metabolism and immunity. In this review, we discuss recent studies that highlight the complex physiological interplay between immune responses and metabolism and give an overview of technological developments that bear the promise of capturing this complexity most directly and comprehensively.
Collapse
Affiliation(s)
- Denis A Mogilenko
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Current affiliation: Department of Medicine, Department of Pathology, Microbiology, and Immunology, and Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| | - Alexey Sergushichev
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
| |
Collapse
|
154
|
Antignano I, Liu Y, Offermann N, Capasso M. Aging microglia. Cell Mol Life Sci 2023; 80:126. [PMID: 37081238 PMCID: PMC10119228 DOI: 10.1007/s00018-023-04775-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/22/2023]
Abstract
Microglia are the tissue-resident macrophage population of the brain, specialized in supporting the CNS environment and protecting it from endogenous and exogenous insults. Nonetheless, their function declines with age, in ways that remain to be fully elucidated. Given the critical role played by microglia in neurodegenerative diseases, a better understanding of the aging microglia phenotype is an essential prerequisite in designing better preventive and therapeutic strategies. In this review, we discuss the most recent literature on microglia in aging, comparing findings in rodent models and human subjects.
Collapse
Affiliation(s)
- Ignazio Antignano
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Yingxiao Liu
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Nina Offermann
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Melania Capasso
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany.
| |
Collapse
|
155
|
Castellani G, Croese T, Peralta Ramos JM, Schwartz M. Transforming the understanding of brain immunity. Science 2023; 380:eabo7649. [PMID: 37023203 DOI: 10.1126/science.abo7649] [Citation(s) in RCA: 153] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Contemporary studies have completely changed the view of brain immunity from envisioning the brain as isolated and inaccessible to peripheral immune cells to an organ in close physical and functional communication with the immune system for its maintenance, function, and repair. Circulating immune cells reside in special niches in the brain's borders, the choroid plexus, meninges, and perivascular spaces, from which they patrol and sense the brain in a remote manner. These niches, together with the meningeal lymphatic system and skull microchannels, provide multiple routes of interaction between the brain and the immune system, in addition to the blood vasculature. In this Review, we describe current ideas about brain immunity and their implications for brain aging, diseases, and immune-based therapeutic approaches.
Collapse
Affiliation(s)
- Giulia Castellani
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Tommaso Croese
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
156
|
Prakash AV, Welliver RR, Mirmire S, Baron S, Hicar MD. Presence of coronary aneurysms during Kawasaki Disease (KD) correlates with lower levels of autoantibodies to both full form and spliced variant of immune regulator Del-1. Immunol Lett 2023; 256-257:34-41. [PMID: 37019289 DOI: 10.1016/j.imlet.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Kawasaki disease (KD), a rare multisystem inflammatory condition that predominantly affects children under six years of age, is the leading cause of childhood-acquired heart disease in developed countries. The pathogenesis is unknown, but studies support that an infectious stimulus triggers an autoimmune reaction in a genetically susceptible child. Recent studies demonstrated an association with autoantibody response to Del-1 (also known as EDIL3) in children with KD. Del-1 is an extracellular matrix protein that is expressed both in macrophages and vascular endothelium. Del-1 has an anti-inflammatory role by preventing leucocyte migration to inflammatory sites. Del-1 has two expression variants and genetic variants of Del-1 have been associated with the risk of intracranial aneurysms. Due to the physiologic plausibility for a role during KD, we chose to assess if autoantibodies against DEL-1 are seen in a larger cohort of children with KD and to assess if responses correlated to aneurysm formation. Contrary to prior findings, in comparison to febrile controls, autoantibodies were not overall higher in children with KD. Elevation in Post-IVIG samples in comparison to pre-IVIG and convalescent samples supports the commonality of anti-Del-1 antibodies. Autoantibodies were notably lower in children with KD who had coronary Z score elevations in comparison to those who did not.
Collapse
Affiliation(s)
- Aviraag Vijaya Prakash
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| | - R Ross Welliver
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| | - Sanjiti Mirmire
- Houston Methodist Hospital, Department of Neurology, Houston, Texas.
| | - Sarah Baron
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| | - Mark D Hicar
- Jacobs School of Medicine and Public Health, Department of Pediatrics, University at Buffalo, Buffalo, New York.
| |
Collapse
|
157
|
Rada CC, Yuki K, Ding J, Kuo CJ. Regulation of the Blood-Brain Barrier in Health and Disease. Cold Spring Harb Perspect Med 2023; 13:a041191. [PMID: 36987582 PMCID: PMC10691497 DOI: 10.1101/cshperspect.a041191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The neurovascular unit is a dynamic microenvironment with tightly controlled signaling and transport coordinated by the blood-brain barrier (BBB). A properly functioning BBB allows sufficient movement of ions and macromolecules to meet the high metabolic demand of the central nervous system (CNS), while protecting the brain from pathogenic and noxious insults. This review describes the main cell types comprising the BBB and unique molecular signatures of these cells. Additionally, major signaling pathways for BBB development and maintenance are highlighted. Finally, we describe the pathophysiology of BBB diseases, their relationship to barrier dysfunction, and identify avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Cara C Rada
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Jie Ding
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
158
|
Fingelkurts AA, Fingelkurts AA. Turning Back the Clock: A Retrospective Single-Blind Study on Brain Age Change in Response to Nutraceuticals Supplementation vs. Lifestyle Modifications. Brain Sci 2023; 13:520. [PMID: 36979330 PMCID: PMC10046544 DOI: 10.3390/brainsci13030520] [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: 02/20/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND There is a growing consensus that chronological age (CA) is not an accurate indicator of the aging process and that biological age (BA) instead is a better measure of an individual's risk of age-related outcomes and a more accurate predictor of mortality than actual CA. In this context, BA measures the "true" age, which is an integrated result of an individual's level of damage accumulation across all levels of biological organization, along with preserved resources. The BA is plastic and depends upon epigenetics. Brain state is an important factor contributing to health- and lifespan. METHODS AND OBJECTIVE Quantitative electroencephalography (qEEG)-derived brain BA (BBA) is a suitable and promising measure of brain aging. In the present study, we aimed to show that BBA can be decelerated or even reversed in humans (N = 89) by using customized programs of nutraceutical compounds or lifestyle changes (mean duration = 13 months). RESULTS We observed that BBA was younger than CA in both groups at the end of the intervention. Furthermore, the BBA of the participants in the nutraceuticals group was 2.83 years younger at the endpoint of the intervention compared with their BBA score at the beginning of the intervention, while the BBA of the participants in the lifestyle group was only 0.02 years younger at the end of the intervention. These results were accompanied by improvements in mental-physical health comorbidities in both groups. The pre-intervention BBA score and the sex of the participants were considered confounding factors and analyzed separately. CONCLUSIONS Overall, the obtained results support the feasibility of the goal of this study and also provide the first robust evidence that halting and reversal of brain aging are possible in humans within a reasonable (practical) timeframe of approximately one year.
Collapse
|
159
|
Chen Y, Hao X, Li M, Tian Z, Cheng M. UGRP1-modulated MARCO + alveolar macrophages contribute to age-related lung fibrosis. Immun Ageing 2023; 20:14. [PMID: 36934284 PMCID: PMC10024420 DOI: 10.1186/s12979-023-00338-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/08/2023] [Indexed: 03/19/2023]
Abstract
The aging lungs are vulnerable to chronic pulmonary diseases; however, the underlying mechanisms are not well understood. In this study, we compared the aging lungs of 20-24-month-old mice with the young of 10-16-week-old mice, and found that aging airway epithelial cells significantly upregulated the expression of uteroglobin-related protein 1 (UGRP1), which was responsible for the higher levels of CCL6 in the aging lungs. Alveolar macrophages (AMs) changed intrinsically with aging, exhibiting a decrease in cell number and altered gene expression. Using terminal differentiation trajectories, a population of MARCO+ AMs with the ability to produce CCL6 was identified in the aging lungs. Upregulated UGRP1was demonstrated to modulate CCL6 production of AMs in the UGRP1-MARCO pair in vivo and in vitro. Furthermore, MARCO+ AMs aggravated bleomycin-induced pulmonary fibrosis in a CCL6-dependent manner in the aged mice, and blocking MARCO or neutralizing CCL6 significantly inhibited pulmonary fibrosis, similar to the depletion of AMs. The age-related upregulation of UGRP1 and MARCO+ AMs, involved in the progression of lung fibrosis, was also observed in human lung tissues. Thus, UGRP1 modulated MARCO+ AMs regarding the age-related lung fibrosis in a CCL6-dependent manner, which is key to establishing optimal targeting for the aging population.
Collapse
Affiliation(s)
- Yongyan Chen
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, China.
| | - Xiaolei Hao
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, China
| | - Ming Li
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, China
- Cancer Immunotherapy Center, the First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, 230001, China
| | - Min Cheng
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, China.
- Cancer Immunotherapy Center, the First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, 230001, China.
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, 230001, China.
| |
Collapse
|
160
|
Henn RE, Guo K, Elzinga SE, Noureldein MH, Mendelson FE, Hayes JM, Rigan DM, Savelieff MG, Hur J, Feldman EL. Single-cell RNA sequencing identifies hippocampal microglial dysregulation in diet-induced obesity. iScience 2023; 26:106164. [PMID: 36915697 PMCID: PMC10006681 DOI: 10.1016/j.isci.2023.106164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/23/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Obesity is a growing global concern in adults and youth with a parallel rise in associated complications, including cognitive impairment. Obesity induces brain inflammation and activates microglia, which contribute to cognitive impairment by aberrantly phagocytosing synaptic spines. Local and systemic signals, such as inflammatory cytokines and metabolites likely participate in obesity-induced microglial activation. However, the precise mechanisms mediating microglial activation during obesity remain incompletely understood. Herein, we leveraged our mouse model of high-fat diet (HFD)-induced obesity, which mirrors human obesity, and develops hippocampal-dependent cognitive impairment. We assessed hippocampal microglial activation by morphological and single-cell transcriptomic analysis to evaluate this heterogeneous, functionally diverse, and dynamic class of cells over time after 1 and 3 months of HFD. HFD altered cell-to-cell communication, particularly immune modulation and cellular adhesion signaling, and induced a differential gene expression signature of protein processing in the endoplasmic reticulum in a time-dependent manner.
Collapse
Affiliation(s)
- Rosemary E. Henn
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Sarah E. Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Mohamed H. Noureldein
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Faye E. Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Diana M. Rigan
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Masha G. Savelieff
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
161
|
Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications. Signal Transduct Target Ther 2023; 8:114. [PMID: 36918543 PMCID: PMC10015017 DOI: 10.1038/s41392-023-01378-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Cardiac aging is evident by a reduction in function which subsequently contributes to heart failure. The metabolic microenvironment has been identified as a hallmark of malignancy, but recent studies have shed light on its role in cardiovascular diseases (CVDs). Various metabolic pathways in cardiomyocytes and noncardiomyocytes determine cellular senescence in the aging heart. Metabolic alteration is a common process throughout cardiac degeneration. Importantly, the involvement of cellular senescence in cardiac injuries, including heart failure and myocardial ischemia and infarction, has been reported. However, metabolic complexity among human aging hearts hinders the development of strategies that targets metabolic susceptibility. Advances over the past decade have linked cellular senescence and function with their metabolic reprogramming pathway in cardiac aging, including autophagy, oxidative stress, epigenetic modifications, chronic inflammation, and myocyte systolic phenotype regulation. In addition, metabolic status is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and chronic inflammation. However, further elucidation of the metabolism involvement in cardiac degeneration is still needed. Thus, deciphering the mechanisms underlying how metabolic reprogramming impacts cardiac aging is thought to contribute to the novel interventions to protect or even restore cardiac function in aging hearts. Here, we summarize emerging concepts about metabolic landscapes of cardiac aging, with specific focuses on why metabolic profile alters during cardiac degeneration and how we could utilize the current knowledge to improve the management of cardiac aging.
Collapse
|
162
|
Suzzi S, Croese T, Ravid A, Gold O, Clark AR, Medina S, Kitsberg D, Adam M, Vernon KA, Kohnert E, Shapira I, Malitsky S, Itkin M, Brandis A, Mehlman T, Salame TM, Colaiuta SP, Cahalon L, Slyper M, Greka A, Habib N, Schwartz M. N-acetylneuraminic acid links immune exhaustion and accelerated memory deficit in diet-induced obese Alzheimer's disease mouse model. Nat Commun 2023; 14:1293. [PMID: 36894557 PMCID: PMC9998639 DOI: 10.1038/s41467-023-36759-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
Systemic immunity supports lifelong brain function. Obesity posits a chronic burden on systemic immunity. Independently, obesity was shown as a risk factor for Alzheimer's disease (AD). Here we show that high-fat obesogenic diet accelerated recognition-memory impairment in an AD mouse model (5xFAD). In obese 5xFAD mice, hippocampal cells displayed only minor diet-related transcriptional changes, whereas the splenic immune landscape exhibited aging-like CD4+ T-cell deregulation. Following plasma metabolite profiling, we identified free N-acetylneuraminic acid (NANA), the predominant sialic acid, as the metabolite linking recognition-memory impairment to increased splenic immune-suppressive cells in mice. Single-nucleus RNA-sequencing revealed mouse visceral adipose macrophages as a potential source of NANA. In vitro, NANA reduced CD4+ T-cell proliferation, tested in both mouse and human. In vivo, NANA administration to standard diet-fed mice recapitulated high-fat diet effects on CD4+ T cells and accelerated recognition-memory impairment in 5xFAD mice. We suggest that obesity accelerates disease manifestation in a mouse model of AD via systemic immune exhaustion.
Collapse
Grants
- R01 DK095045 NIDDK NIH HHS
- R01 DK099465 NIDDK NIH HHS
- the Vera and John Schwartz Family Center for Metabolic Biology.
- the National Institutes of Health (NIH) grants DK095045 and DK099465, the Cure Alzheimer’s Fund, the Chan Zuckerberg Foundation, and the Carlos Slim Foundation.
- the Israel Science Foundation (ISF) research grant no. 1709/19, the European Research Council grant 853409, the MOST-IL-China research grant no. 3-15687, and the Myers Foundation. N.H. holds the Goren-Khazzam chair in neuroscience.
- the Advanced European Research Council grants 232835 and 741744, the European Seventh Framework Program HEALTH-2011 (279017), the Israel Science Foundation (ISF)-research grant no. 991/16, the ISF-Legacy Heritage Bio-medical Science Partnership research grant no. 1354/15, and the Thompson Foundation and Adelis Foundation.
Collapse
Affiliation(s)
- Stefano Suzzi
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel.
| | - Tommaso Croese
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Adi Ravid
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Or Gold
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Abbe R Clark
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sedi Medina
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Daniel Kitsberg
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Miriam Adam
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Katherine A Vernon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eva Kohnert
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Inbar Shapira
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Sergey Malitsky
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Maxim Itkin
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Alexander Brandis
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Tevie Mehlman
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Tomer M Salame
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Sarah P Colaiuta
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Liora Cahalon
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Michal Slyper
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Naomi Habib
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel.
| | - Michal Schwartz
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel.
| |
Collapse
|
163
|
Yin F. Lipid metabolism and Alzheimer's disease: clinical evidence, mechanistic link and therapeutic promise. FEBS J 2023; 290:1420-1453. [PMID: 34997690 PMCID: PMC9259766 DOI: 10.1111/febs.16344] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/14/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disorder with multifactorial etiology, intersecting genetic and environmental risk factors, and a lack of disease-modifying therapeutics. While the abnormal accumulation of lipids was described in the very first report of AD neuropathology, it was not until recent decades that lipid dyshomeostasis became a focus of AD research. Clinically, lipidomic and metabolomic studies have consistently shown alterations in the levels of various lipid classes emerging in early stages of AD brains. Mechanistically, decades of discovery research have revealed multifaceted interactions between lipid metabolism and key AD pathogenic mechanisms including amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. In the present review, converging evidence defining lipid dyshomeostasis in AD is summarized, followed by discussions on mechanisms by which lipid metabolism contributes to pathogenesis and modifies disease risk. Furthermore, lipid-targeting therapeutic strategies, and the modification of their efficacy by disease stage, ApoE status, and metabolic and vascular profiles, are reviewed.
Collapse
Affiliation(s)
- Fei Yin
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA.,Department of Pharmacology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA.,Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
164
|
Zhang Q, Song Q, Yu R, Wang A, Jiang G, Huang Y, Chen J, Xu J, Wang D, Chen H, Gao X. Nano-Brake Halts Mitochondrial Dysfunction Cascade to Alleviate Neuropathology and Rescue Alzheimer's Cognitive Deficits. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204596. [PMID: 36703613 PMCID: PMC9982524 DOI: 10.1002/advs.202204596] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/02/2023] [Indexed: 06/18/2023]
Abstract
Mitochondrial dysfunction has been recognized as the key pathogenesis of most neurodegenerative diseases including Alzheimer's disease (AD). The dysregulation of mitochondrial calcium ion (Ca2+ ) homeostasis and the mitochondrial permeability transition pore (mPTP), is a critical upstream signaling pathway that contributes to the mitochondrial dysfunction cascade in AD pathogenesis. Herein, a "two-hit braking" therapeutic strategy to synergistically halt mitochondrial Ca2+ overload and mPTP opening to put the mitochondrial dysfunction cascade on a brake is proposed. To achieve this goal, magnesium ion (Mg2+ ), a natural Ca2+ antagonist, and siRNA to the central mPTP regulator cyclophilin D (CypD), are co-encapsulated into the designed nano-brake; A matrix metalloproteinase 9 (MMP9) activatable cell-penetrating peptide (MAP) is anchored on the surface of nano-brake to overcome the blood-brain barrier (BBB) and realize targeted delivery to the mitochondrial dysfunction cells of the brain. Nano-brake treatment efficiently halts the mitochondrial dysfunction cascade in the cerebrovascular endothelial cells, neurons, and microglia and powerfully alleviates AD neuropathology and rescues cognitive deficits. These findings collectively demonstrate the potential of advanced design of nanotherapeutics to halt the key upstream signaling pathways of mitochondrial dysfunction to provide a powerful strategy for AD modifying therapy.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Qingxiang Song
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Renhe Yu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Antian Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Gan Jiang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yukun Huang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Jun Chen
- School of PharmacyShanghai Pudong Hospital & Department of PharmaceuticsFudan UniversityLane 826, Zhangheng RoadShanghai201203China
| | - Jianrong Xu
- Academy of Integrative MedicineShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201203China
| | - Dayuan Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Hongzhuan Chen
- Institute of Interdisciplinary Integrative Biomedical ResearchShuguang HospitalShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201203China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| |
Collapse
|
165
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
166
|
Li W, Li S, Shang Y, Zhuang W, Yan G, Chen Z, Lyu J. Associations between dietary and blood inflammatory indices and their effects on cognitive function in elderly Americans. Front Neurosci 2023; 17:1117056. [PMID: 36895419 PMCID: PMC9989299 DOI: 10.3389/fnins.2023.1117056] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
OBJECTIVE To determine the correlations between dietary and blood inflammation indices in elderly Americans and their effects on cognitive function. METHODS This research extracted data from the 2011-2014 National Health and Nutrition Examination Survey for 2,479 patients who were ≥60 years old. Cognitive function was assessed as a composite cognitive function score (Z-score) calculated from the results of the Consortium to Establish a Registry for Alzheimer's Disease Word Learning and Delayed Recall tests, the Animal Fluency test, and the Digit Symbol Substitution Test. We used a dietary inflammatory index (DII) calculated from 28 food components to represent the dietary inflammation profile. Blood inflammation indicators included the white blood cell count (WBC), neutrophil count (NE), lymphocyte count (Lym), neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), neutrophil-albumin ratio (NAR), systemic immune-inflammation index [SII, calculated as (peripheral platelet count) × NE/Lym], and systemic inflammatory response index [SIRI, calculated as (monocyte count) × NE/Lym]. WBC, NE, Lym, NLR, PLR, NAR, SII, SIRI, and DII were initially treated as continuous variables. For logistic regression, WBC, NE, Lym, NLR, PLR, NAR, SII, and SIRI were divided into quartile groups, and DII was divided into tertile groups. RESULTS After adjusting for covariates, WBC, NE, NLR, NAR, SII, SIRI, and DII scores were markedly higher in the cognitively impaired group than in the normal group (p < 0.05). DII was negatively correlated with the Z-score when combined with WBC, NE, and NAR (p < 0.05). After adjusting for all covariates, DII was positively correlated with SII in people with cognitive impairment (p < 0.05). Higher DII with NLR, NAR, SII, and SIRI all increased the risk of cognitive impairment (p < 0.05). CONCLUSION DII was positively correlated with blood inflammation indicators, and higher DII and blood inflammation indicators increased the risk of developing cognitive impairment.
Collapse
Affiliation(s)
- Wanyue Li
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shuna Li
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yaru Shang
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Weisheng Zhuang
- Department of Rehabilitation, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guoqiang Yan
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhuoming Chen
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, China
| |
Collapse
|
167
|
Zhao Y, Li H, Guo Q, Hui H. Multiple characteristic alterations and available therapeutic strategies of cellular senescence. J Zhejiang Univ Sci B 2023; 24:101-114. [PMID: 36751697 PMCID: PMC9936135 DOI: 10.1631/jzus.b2200178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Given its state of stable proliferative inhibition, cellular senescence is primarily depicted as a critical mechanism by which organisms delay the progression of carcinogenesis. Cells undergoing senescence are often associated with the alteration of a series of specific features and functions, such as metabolic shifts, stemness induction, and microenvironment remodeling. However, recent research has revealed more complexity associated with senescence, including adverse effects on both physiological and pathological processes. How organisms evade these harmful consequences and survive has become an urgent research issue. Several therapeutic strategies targeting senescence, including senolytics, senomorphics, immunotherapy, and function restoration, have achieved initial success in certain scenarios. In this review, we describe in detail the characteristic changes associated with cellular senescence and summarize currently available countermeasures.
Collapse
Affiliation(s)
- Yunzi Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009 China
| | - Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009 China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009 China
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
168
|
Varvel NH, Amaradhi R, Espinosa-Garcia C, Duddy S, Franklin R, Banik A, Alemán-Ruiz C, Blackmer-Raynolds L, Wang W, Honore T, Ganesh T, Dingledine R. Preclinical development of an EP2 antagonist for post-seizure cognitive deficits. Neuropharmacology 2023; 224:109356. [PMID: 36460083 PMCID: PMC9894535 DOI: 10.1016/j.neuropharm.2022.109356] [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: 09/30/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022]
Abstract
Cognitive comorbidities can substantially reduce quality of life in people with epilepsy. Inflammation is a component of all chronic diseases including epilepsy, as well as acute events like status epilepticus (SE). Neuroinflammation is the consequence of several broad signaling cascades including cyclooxygenase-2 (COX-2)-associated pathways. Activation of the EP2 receptor for prostaglandin E2 appears responsible for blood-brain barrier leakage and much of the inflammatory reaction, neuronal injury and cognitive deficit that follows seizure-provoked COX-2 induction in brain. Here we show that brief exposure of mice to TG11-77, a potent, selective, orally available and brain permeant EP2 antagonist, eliminates the profound cognitive deficit in Y-maze performance after SE and reduces delayed mortality and microgliosis, with a minimum effective i.p. dose (as free base) of 8.8 mg/kg. All in vitro studies required to submit an investigational new drug (IND) application for TG11-77 have been completed, and non-GLP dose range-finding toxicology in the rat identified no overt, organ or histopathology signs of toxicity after 7 days of oral administration at 1000 mg/kg/day. Plasma exposure in the rat was dose-linear between 15 and 1000 mg/kg dosing. TG11-77 thus appears poised to continue development towards the initial clinical test of the hypothesis that EP2 receptor modulation after SE can provide the first preventive treatment for one of the chief comorbidities of epilepsy.
Collapse
Affiliation(s)
- Nicholas H Varvel
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Claudia Espinosa-Garcia
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Steven Duddy
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Ronald Franklin
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Avijit Banik
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Carlos Alemán-Ruiz
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Lisa Blackmer-Raynolds
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Tage Honore
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia.
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia.
| |
Collapse
|
169
|
Leyder E, Suresh P, Jun R, Overbey K, Banerjee T, Melnikova T, Savonenko A. Depression-related phenotypes at early stages of Aβ and tau accumulation in inducible Alzheimer's disease mouse model: Task-oriented and concept-driven interpretations. Behav Brain Res 2023; 438:114187. [PMID: 36343696 DOI: 10.1016/j.bbr.2022.114187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 10/16/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Depression is highly prevalent in Alzheimer Disease (AD); however, there is paucity of studies that focus specifically on the assessment of depression-relevant phenotypes in AD mouse models. Conditional doxycycline-dependent transgenic mouse models reproducing amyloidosis (TetOffAPPsi) and/or tau (TetOffTauP301L) pathology starting at middle age (6 months) were used in this study. As AD patients can experience depressive symptoms relatively early in disease, testing was conducted at early, pre-pathology stages of Aβ and/or tau accumulation (starting from 45 days of transgenes expression). Tau-related differences were detected in the Novelty Suppressed Feeding task (NSF), whereas APP-related differences were observed predominantly in measures of the Open Field (OF) and Forced Swim tasks (FST). Effects of combined production of Aβ and tau were detected in immobility during the 1st half of the Tail Suspension task (TST). These data demonstrate that results from different tasks are difficult to reconcile using task/variable-centered interpretations in which a single task/variable is assigned an ad-hoc meaning relevant to depression. An alternative, concept-oriented, approach is based on multiple variables/tests, with an understanding of their possible inter-dependence and utilization of statistical approaches that handle correlated data sets. The existence of strong correlations within and between some of the tasks supported utilization of factor analyses (FA). FA explained a similar amount of variability across the genotypes (∼80%) and identified two factors stable across genotypes and representing motor activity and anxiety measures in OF. In contrast, variables related to FST, TST, and NSFT did not demonstrate a structure of factor loadings that would support the existence of a single integral factor of "depressive state" measured by these tasks. In addition, factor loadings varied between genotypes, indicating that genotype-specific between-task correlations need to be considered for interpretations of findings in any single task. In general, this study demonstrates that utilization of multiple tasks to characterize behavioral phenotypes, an approach that is finally gaining more widespread adoption, requires a step of data integration across different behavioral tests for appropriate interpretations.
Collapse
Affiliation(s)
- Erica Leyder
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Prakul Suresh
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Rachel Jun
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Katherine Overbey
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Tirtho Banerjee
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Tatiana Melnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | - Alena Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
170
|
Linghui D, Simin Y, Zilong Z, Yuxiao L, Shi Q, Birong D. The relationship between serum klotho and cognitive performance in a nationally representative sample of US adults. Front Aging Neurosci 2023; 15:1053390. [PMID: 36819720 PMCID: PMC9932504 DOI: 10.3389/fnagi.2023.1053390] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 01/05/2023] [Indexed: 02/05/2023] Open
Abstract
Purpose Aging is the primary risk factor for cognitive decline. Serum klotho, as an anti-aging protein, may be involved in cognitive decline. Thus, we aim to explorer the correlation between serum klotho and cognitive performance among an older adult population in the United States. Methods We performed a cross-sectional study using data from NHANES 2011-2014. Serum klotho was analyzed by ELISA. Cognitive function was measured by Establish a Registry for Alzheimer's Disease (CERAD) test, Animal Fluency test and Digit Symbol Substitution Test (DSST) score. The relationship between serum klotho and cognition was analyzed by a multivariable regression model. Results A total of 2,171 participants aged 60-79 years were included. Median serum klotho concentration was 851.52 pg./ml (SD = 294.07). We also categorized serum klotho concentrates into quartiles. After fully adjusting pertinent variables, compared to those with lowest klotho levels (206.3-658.4 pg./ml), individuals with highest klotho concentrates (983.3-3,456 pg./ml) had a higher CERAD score [β (95%CI): 0.97 (0.25, 1.69) p = 0.008] and DSST score [β (95%CI): 1.86 (0.25, 3.47), p = 0.024]. Conclusion Our findings indicated that, among the general population of American older adults, serum klotho concentrates may serve as a marker of cognitive health. The benefits of klotho on aging process and neurodegenerative disorders should be paid more attention.
Collapse
Affiliation(s)
- Deng Linghui
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Simin
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Zhang Zilong
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Li Yuxiao
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qiu Shi
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China,*Correspondence: Qiu Shi, ✉
| | - Dong Birong
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China,Dong Birong, ✉
| |
Collapse
|
171
|
Tuohy MC, Hillman EMC, Marshall R, Agalliu D. The age-dependent immune response to ischemic stroke. Curr Opin Neurobiol 2023; 78:102670. [PMID: 36586305 PMCID: PMC9845177 DOI: 10.1016/j.conb.2022.102670] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/31/2022]
Abstract
Stroke is a devastating cause of global morbidity and mortality. Ischemic brain injury triggers a profound local and systemic immune response that participates in stroke pathophysiology. In turn, this immune response has emerged as a potential therapeutic target. In order to maximize its therapeutic potential, it is critical to understand how the immune response to ischemic brain injury is affected by age - the strongest non-modifiable risk factor for stroke. The development of multi-omics and single-cell technologies has provided a more comprehensive characterization of transcriptional and cellular changes that occur during aging. In this review, we summarize recent advances in our understanding of how age-related immune alterations shape differential stroke outcomes in older versus younger organisms, highlighting studies in both experimental mouse models and patient cohorts. Wherever possible, we emphasize outstanding questions that present important avenues for future investigation with therapeutic value for the aging population.
Collapse
Affiliation(s)
- Mary Claire Tuohy
- Doctoral Program in Neurobiology and Behavior, Columbia University Irving Medical Center, New York, NY, 10032, USA; Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA; Zuckerman Mind Brain Behavior Institute and Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Elizabeth M C Hillman
- Zuckerman Mind Brain Behavior Institute and Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA. https://twitter.com/HillmanLab
| | - Randolph Marshall
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Dritan Agalliu
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
172
|
Jin M, Cai SQ. Mechanisms Underlying Brain Aging Under Normal and Pathological Conditions. Neurosci Bull 2023; 39:303-314. [PMID: 36437436 PMCID: PMC9905409 DOI: 10.1007/s12264-022-00969-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022] Open
Abstract
Aging is a major risk factor for many human diseases, including cognitive impairment, which affects a large population of the elderly. In the past few decades, our understanding of the molecular and cellular mechanisms underlying the changes associated with aging and age-related diseases has expanded greatly, shedding light on the potential role of these changes in cognitive impairment. In this article, we review recent advances in understanding of the mechanisms underlying brain aging under normal and pathological conditions, compare their similarities and differences, discuss the causative and adaptive mechanisms of brain aging, and finally attempt to find some rules to guide us on how to promote healthy aging and prevent age-related diseases.
Collapse
Affiliation(s)
- Menglong Jin
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shi-Qing Cai
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
173
|
Wang Y, Yang M, Zhang J, Ren J, Liu N, Liu B, Lu L, Yang B. S-Doped carbonized polymer dots inhibit early myocardial fibrosis by regulating mitochondrial function. Biomater Sci 2023; 11:894-907. [PMID: 36524407 DOI: 10.1039/d2bm00578f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Myocardial fibrosis (MF) is a critical pathological lesion in the progression of various acute and chronic cardiovascular diseases. However, there is still a lack of clinically effective drugs and treatments for MF therapies. Herein, for the first time, we developed fluorescent sulfur-doped carbonized polymer dots (S-CPDs) as new nano-antioxidants to reduce the cardiomyocyte damage caused by reactive oxygen species (ROS) in the early stage of fibrotic lesions. In vitro results suggested that the pre-protection of S-CPDs significantly increased the survival rate of H9c2 cells under severe oxidative stress, inhibited the isoproterenol (ISO)-induced hypertrophy of myocardial cells through improving the content of mitochondria related proteins and adenosine triphosphate (ATP) in cells. Moreover, S-CPD administration could effectively decrease cardiac hypertrophy and promote heart function in MF rat models. The rapid internalization, high biocompatibility and fluorescence imaging potential of S-CPDs revealed their promising application prospects in the diagnoses and treatments of cardiovascular diseases.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Mingxi Yang
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China. .,State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P.R. China.
| | - Jiayi Zhang
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Jingyan Ren
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Laijin Lu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P.R. China.
| |
Collapse
|
174
|
Chen HY, Zhao Y, Xie YZ. Immunosenescence of brain accelerates Alzheimer's disease progression. Rev Neurosci 2023; 34:85-101. [PMID: 35791032 DOI: 10.1515/revneuro-2022-0021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/04/2022] [Indexed: 01/07/2023]
Abstract
Most of Alzheimer's disease (AD) cases are sporadic and occur after age 65. With prolonged life expectancy and general population aging, AD is becoming a significant public health concern. The immune system supports brain development, plasticity, and homeostasis, yet it is particularly vulnerable to aging-related changes. Aging of the immune system, called immunosenescence, is the multifaceted remodeling of the immune system during aging. Immunosenescence is a contributing factor to various age-related diseases, including AD. Age-related changes in brain immune cell phenotype and function, crosstalk between immune cells and neural cells, and neuroinflammation work together to promote neurodegeneration and age-related cognitive impairment. Although numerous studies have confirmed the correlation between systemic immune changes and AD, few studies focus on the immune state of brain microenvironment in aging and AD. This review mainly addresses the changes of brain immune microenvironment in aging and AD. Specifically, we delineate how various aspects of the brain immune microenvironment, including immune gateways, immune cells, and molecules, and the interplay between immune cells and neural cells, accelerate AD pathogenesis during aging. We also propose a theoretical framework of therapeutic strategies selectively targeting the different mechanisms to restore brain immune homeostasis.
Collapse
Affiliation(s)
- Hou-Yu Chen
- Department of Abdominal Surgery, Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangdong 510095, China
| | - Yan Zhao
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Yong-Zhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
175
|
Biological Aging in People Living with HIV on Successful Antiretroviral Therapy: Do They Age Faster? Curr HIV/AIDS Rep 2023; 20:42-50. [PMID: 36695947 PMCID: PMC10102129 DOI: 10.1007/s11904-023-00646-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2023] [Indexed: 01/26/2023]
Abstract
PURPOSE OF REVIEW In the absence of a prophylactic/therapeutic vaccine or cure, the most amazing achievement in the battle against HIV was the discovery of effective, well-tolerated combination antiretroviral therapy (cART). The primary research question remains whether PLWH on prolonged successful therapy has accelerated, premature, or accentuated biological aging. In this review, we discuss the current understanding of the immunometabolic profile in PLWH, potentially associated with biological aging, and a better understanding of the mechanisms and temporal dynamics of biological aging in PLWH. RECENT FINDINGS Biological aging, defined by the epigenetic alterations analyzed by the DNA methylation pattern, has been reported in PLWH with cART that points towards epigenetic age acceleration. The hastened development of specific clinical geriatric syndromes like cardiovascular diseases, metabolic syndrome, cancers, liver diseases, neurocognitive diseases, persistent low-grade inflammation, and a shift toward glutamate metabolism in PLWH may potentiate a metabolic profile at-risk for accelerated aging.
Collapse
|
176
|
The role of ApoE-mediated microglial lipid metabolism in brain aging and disease. IMMUNOMETABOLISM (COBHAM (SURREY, ENGLAND)) 2023; 5:e00018. [PMID: 36710921 PMCID: PMC9869962 DOI: 10.1097/in9.0000000000000018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/20/2022] [Indexed: 01/31/2023]
Abstract
Microglia are a unique population of immune cells resident in the brain that integrate complex signals and dynamically change phenotypes in response to the brain microenvironment. In recent years, single-cell sequencing analyses have revealed profound cellular heterogeneity and context-specific transcriptional plasticity of microglia during brain development, aging, and disease. Emerging evidence suggests that microglia adapt phenotypic plasticity by flexibly reprogramming cellular metabolism to fulfill distinct immune functions. The control of lipid metabolism is central to the appropriate function and homeostasis of the brain. Microglial lipid metabolism regulated by apolipoprotein E (ApoE), a crucial lipid transporter in the brain, has emerged as a critical player in regulating neuroinflammation. The ApoE gene allelic variant, ε4, is associated with a greater risk for neurodegenerative diseases. In this review, we explore novel discoveries in microglial lipid metabolism mediated by ApoE. We elaborate on the functional impact of perturbed microglial lipid metabolism on the underlying pathogenesis of brain aging and disease.
Collapse
|
177
|
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: An expanding universe. Cell 2023; 186:243-278. [PMID: 36599349 DOI: 10.1016/j.cell.2022.11.001] [Citation(s) in RCA: 2170] [Impact Index Per Article: 1085.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/19/2022] [Accepted: 11/01/2022] [Indexed: 01/05/2023]
Abstract
Aging is driven by hallmarks fulfilling the following three premises: (1) their age-associated manifestation, (2) the acceleration of aging by experimentally accentuating them, and (3) the opportunity to decelerate, stop, or reverse aging by therapeutic interventions on them. We propose the following twelve hallmarks of aging: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. These hallmarks are interconnected among each other, as well as to the recently proposed hallmarks of health, which include organizational features of spatial compartmentalization, maintenance of homeostasis, and adequate responses to stress.
Collapse
Affiliation(s)
- Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Linda Partridge
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK; Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain; Altos Labs, Cambridge, UK
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
178
|
Liu ZH, Wang YJ, Bu XL. Alzheimer's disease: targeting the peripheral circulation. Mol Neurodegener 2023; 18:3. [PMID: 36631811 PMCID: PMC9832651 DOI: 10.1186/s13024-023-00594-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Affiliation(s)
- Zhi-Hao Liu
- grid.410570.70000 0004 1760 6682Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China ,grid.414906.e0000 0004 1808 0918Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan-Jiang Wang
- grid.410570.70000 0004 1760 6682Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China ,grid.414906.e0000 0004 1808 0918Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China ,grid.410570.70000 0004 1760 6682Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China ,grid.9227.e0000000119573309Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xian-Le Bu
- grid.410570.70000 0004 1760 6682Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China ,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China ,grid.410570.70000 0004 1760 6682Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| |
Collapse
|
179
|
Liu G, Li T, Yang A, Zhang X, Qi S, Feng W. Knowledge domains and emerging trends of microglia research from 2002 to 2021: A bibliometric analysis and visualization study. Front Aging Neurosci 2023; 14:1057214. [PMID: 36688156 PMCID: PMC9849393 DOI: 10.3389/fnagi.2022.1057214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Background Microglia have been identified for a century. In this period, their ontogeny and functions have come to light thanks to the tireless efforts of scientists. However, numerous documents are being produced, making it challenging for scholars, especially those new to the field, to understand them thoroughly. Therefore, having a reliable method for quickly grasping a field is crucial. Methods We searched and downloaded articles from the Web of Science Core Collection with "microglia" or "microglial" in the title from 2002 to 2021. Eventually, 12,813 articles were located and, using CiteSpace and VOSviewer, the fundamental data, knowledge domains, hot spots, and emerging trends, as well as the influential literature in the field of microglia research, were analyzed. Results Following 2011, microglia publications grew significantly. The two prominent journals are Glia and J Neuroinflamm. The United States and Germany dominated the microglia study. The primary research institutions are Harvard Univ and Univ Freiburg, and the leading authors are Prinz Marco and Kettenmann Helmut. The knowledge domains of microglia include eight directions, namely neuroinflammation, lipopolysaccharide, aging, neuropathic pain, macrophages, Alzheimer's disease, retina, and apoptosis. Microglial phenotype is the focus of research; while RNA-seq, exosome, and glycolysis are emerging topics, a microglial-specific marker is still a hard stone. We also identified 19 influential articles that contributed to the study of microglial origin (Mildner A 2007; Ginhoux F 2010), identity (Butovsky O 2014), homeostasis (Cardona AE 2006; Elmore MRP 2014); microglial function such as surveillance (Nimmerjahn A 2005), movement (Davalos D 2005; Haynes SE 2006), phagocytosis (Simard AR 2006), and synapse pruning (Wake H 2009; Paolicelli RC 2011; Schafer DP 2012; Parkhurst CN 2013); and microglial state/phenotype associated with disease (Keren-Shaul H 2017), as well as 5 review articles represented by Kettenmann H 2011. Conclusion Using bibliometrics, we have investigated the fundamental data, knowledge structure, and dynamic evolution of microglia research over the previous 20 years. We hope this study can provide some inspiration and a reference for researchers studying microglia in neuroscience.
Collapse
Affiliation(s)
- Guangjie Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tianhua Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China,China International Neuroscience Institute (China-INI), Beijing, China
| | - Anming Yang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,*Correspondence: Songtao Qi, ✉
| | - Wenfeng Feng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,Wenfeng Feng, ✉
| |
Collapse
|
180
|
Okahara K, Ohsawa M, Haruta-Tsukamoto A, Miyoshi R, Funahashi H, Fukutani Y, Makita S, Matsuo H, Ishida Y. Frailty Improvement by Multicomponent Drug, Ninjin'Yoeito, in Mild Cognitive Impairment and Mild Alzheimer's Disease Patients: An Open-Label Exploratory Study (FRAMINGO). J Alzheimers Dis Rep 2023; 7:107-117. [PMID: 36891253 PMCID: PMC9986705 DOI: 10.3233/adr-220074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 01/02/2023] [Indexed: 01/25/2023] Open
Abstract
Background Alzheimer's disease (AD) and dementia have increasingly been conceived of as "complex diseases of aging", determined by multiple, simultaneous, interacting pathophysiological processes. The condition known as frailty is a phenotype of aging and its comprehensive pathophysiology is thought to be closely related to the incidence of mild cognitive impairment (MCI) and the exacerbation of dementia. Objective This study aimed to investigate the effect of the multicomponent drug, ninjin'yoeito (NYT), on frailty in MCI and mild AD patients. Methods This study was an open-label trial. A total of 14 patients, including 9 with MCI and 5 with mild AD, were enrolled. Among them, 11 were frail while 3 were prefrail. NYT (6-9 g/day) was administered orally for 24 weeks, and assessments were carried out at baseline (week 0), and at 4, 8, 16, and 24 weeks. Results In the primary endpoint, significant early improvements were observed in the anorexia scores according to the Neuropsychiatric Inventory after four weeks of treatment with NYT. The Cardiovascular Health Study score was significantly improved, and no frailty was observed after 24 weeks. The fatigue visual analog scale scores also significantly improved. The Clinical Dementia Rating and the Montreal Cognitive Assessment scores remained at baseline levels during the NYT treatment period. Conclusion The results suggest that NYT may be effective in the treatment of frailty, especially for anorexia and fatigue, in both MCI and mild AD patients, which would be beneficial for the prognosis of dementia.
Collapse
Affiliation(s)
| | | | - Ayaka Haruta-Tsukamoto
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki-city, Miyazaki, Japan
| | - Ryoei Miyoshi
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki-city, Miyazaki, Japan.,Heartopia Miyoshi Clinic, Miyazaki-city, Miyazaki, Japan
| | - Hideki Funahashi
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki-city, Miyazaki, Japan
| | | | | | - Hisae Matsuo
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki-city, Miyazaki, Japan.,Center for Health Sciences and Counseling, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Yasushi Ishida
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki-city, Miyazaki, Japan
| |
Collapse
|
181
|
Cai Z, He B. Adipose tissue aging: An update on mechanisms and therapeutic strategies. Metabolism 2023; 138:155328. [PMID: 36202221 DOI: 10.1016/j.metabol.2022.155328] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
Aging is a complex biological process characterized by a progressive loss of physiological integrity and increased vulnerability to age-related diseases. Adipose tissue plays central roles in the maintenance of whole-body metabolism homeostasis and has recently attracted significant attention as a biological driver of aging and age-related diseases. Here, we review the most recent advances in our understanding of the molecular and cellular mechanisms underlying age-related decline in adipose tissue function. In particular, we focus on the complex inter-relationship between metabolism, immune, and sympathetic nervous system within adipose tissue during aging. Moreover, we discuss the rejuvenation strategies to delay aging and extend lifespan, including senescent cell ablation (senolytics), dietary intervention, physical exercise, and heterochronic parabiosis. Understanding the pathological mechanisms that underlie adipose tissue aging will be critical for the development of new intervention strategies to slow or reverse aging and age-related diseases.
Collapse
Affiliation(s)
- Zhaohua Cai
- Heart Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ben He
- Heart Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China.
| |
Collapse
|
182
|
McKee CG, Hoffos M, Vecchiarelli HA, Tremblay MÈ. Microglia: A pharmacological target for the treatment of age-related cognitive decline and Alzheimer's disease. Front Pharmacol 2023; 14:1125982. [PMID: 36969855 PMCID: PMC10034122 DOI: 10.3389/fphar.2023.1125982] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/03/2023] [Indexed: 03/29/2023] Open
Abstract
As individuals age, microglia, the resident immune cells of the central nervous system (CNS), become less effective at preserving brain circuits. Increases in microglial inflammatory activity are thought to contribute to age-related declines in cognitive functions and to transitions toward mild cognitive impairment (MCI) and Alzheimer's disease (AD). As microglia possess receptors for communicating with the CNS environment, pharmacological therapies targeting these pathways hold potential for promoting homeostatic microglial functions within the aging CNS. Preclinical and early phase clinical trials investigating the therapeutic effects of pharmacological agents acting on microglia, including reactive oxygen species, TREM2, fractalkine signaling, the complement cascade, and the NLRP3 inflammasome, are currently underway; however, important questions remain unanswered. Current challenges include target selectivity, as many of the signaling pathways are expressed in other cell types. Furthermore, microglia are a heterogenous cell population with transcriptomic, proteomic, and microscopy studies revealing distinct microglial states, whose activities and abundance shift across the lifespan. For example, homeostatic microglia can transform into pathological states characterized by markers of oxidative stress. Selective pharmacological targeting aimed at limiting transitions to pathological states or promoting homeostatic or protective states, could help to avoid potentially harmful off-target effects on beneficial states or other cell types. In this mini-review we cover current microglial pathways of interest for the prevention and treatment of age-related cognitive decline and CNS disorders of aging focusing on MCI and AD. We also discuss the heterogeneity of microglia described in these conditions and how pharmacological agents could target specific microglial states.
Collapse
Affiliation(s)
- Chloe G. McKee
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Madison Hoffos
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
183
|
Long-term potentiation and depression regulatory microRNAs were highlighted in Bisphenol A induced learning and memory impairment by microRNA sequencing and bioinformatics analysis. PLoS One 2023; 18:e0279029. [PMID: 36656826 PMCID: PMC9851566 DOI: 10.1371/journal.pone.0279029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/28/2022] [Indexed: 01/20/2023] Open
Abstract
The mechanisms of Bisphenol A (BPA) induced learning and memory impairment have still not been fully elucidated. MicroRNAs (miRNAs) are endogenous non-coding small RNA molecules involved in the process of toxicant-induced neurotoxicity. To investigate the role of miRNAs in BPA-induced learning and memory impairment, we analyzed the impacts of BPA on miRNA expression profile by high-throughput sequencing in mice hippocampus. Results showed that mice treated with BPA displayed impairments of spatial learning and memory and changes in the expression of miRNAs in the hippocampus. Seventeen miRNAs were significantly differentially expressed after BPA exposure, of these, 13 and 4 miRNAs were up- and downregulated, respectively. Bioinformatic analysis of Gene Ontology (GO) and pathway suggests that BPA exposure significantly triggered transcriptional changes of miRNAs associated with learning and memory; the top five affected pathways involved in impairment of learning and memory are: 1) Long-term depression (LTD); 2) Thyroid hormone synthesis; 3) GnRH signaling pathway; 4) Long-term potentiation (LTP); 5) Serotonergic synapse. Eight BPA-responsive differentially expressed miRNAs regulating LTP and LTD were further screened to validate the miRNA sequencing data using Real-Time PCR. The deregulation expression levels of proteins of five target genes (CaMKII, MEK1/2, IP3R, AMPAR1 and PLCβ4) were investigated via western blot, for further verifying the results of gene target analysis. Our results showed that LTP and LTD related miRNAs and their targets could contribute to BPA-induced impairment of learning and memory. This study provides valuable information for novel miRNA biomarkers to detect changes in impairment of learning and memory induced by BPA exposure.
Collapse
|
184
|
Zhang P, Pan S, Yuan S, Shang Y, Shu H. Abnormal glucose metabolism in virus associated sepsis. Front Cell Infect Microbiol 2023; 13:1120769. [PMID: 37124033 PMCID: PMC10130199 DOI: 10.3389/fcimb.2023.1120769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Sepsis is identified as a potentially lethal organ impairment triggered by an inadequate host reaction to infection (Sepsis-3). Viral sepsis is a potentially deadly organ impairment state caused by the host's inappropriate reaction to a viral infection. However, when a viral infection occurs, the metabolism of the infected cell undergoes a variety of changes that cause the host to respond to the infection. But, until now, little has been known about the challenges faced by cellular metabolic alterations that occur during viral infection and how these changes modulate infection. This study concentrates on the alterations in glucose metabolism during viral sepsis and their impact on viral infection, with a view to exploring new potential therapeutic targets for viral sepsis.
Collapse
Affiliation(s)
| | | | | | - You Shang
- *Correspondence: Huaqing Shu, ; You Shang,
| | | |
Collapse
|
185
|
Gao S, Jiang Y, Chen Z, Zhao X, Gu J, Wu H, Liao Y, Sun H, Wang J, Chen W. Metabolic Reprogramming of Microglia in Sepsis-Associated Encephalopathy: Insights from Neuroinflammation. Curr Neuropharmacol 2023; 21:1992-2005. [PMID: 36529923 PMCID: PMC10514522 DOI: 10.2174/1570159x21666221216162606] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction caused by sepsis that manifests as a range of brain dysfunctions from delirium to coma. It is a relatively common complication of sepsis associated with poor patient prognosis and mortality. The pathogenesis of SAE involves neuroinflammatory responses, neurotransmitter dysfunction, blood-brain barrier (BBB) disruption, abnormal blood flow regulation, etc. Neuroinflammation caused by hyperactivation of microglia is considered to be a key factor in disease development, which can cause a series of chain reactions, including BBB disruption and oxidative stress. Metabolic reprogramming has been found to play a central role in microglial activation and executive functions. In this review, we describe the pivotal role of energy metabolism in microglial activation and functional execution and demonstrate that the regulation of microglial metabolic reprogramming might be crucial in the development of clinical therapeutics for neuroinflammatory diseases like SAE.
Collapse
Affiliation(s)
- Shenjia Gao
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yi Jiang
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhaoyuan Chen
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xiaoqiang Zhao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Jiahui Gu
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Han Wu
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yun Liao
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Hao Sun
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jun Wang
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Fudan Zhangjiang Institute, Shanghai, 201203, China
| |
Collapse
|
186
|
Chang ZS, He ZM, Xia JB. FoxO3 Regulates the Progress and Development of Aging and Aging-Related Diseases. Curr Mol Med 2023; 23:991-1006. [PMID: 36239722 DOI: 10.2174/1566524023666221014140817] [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: 06/20/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/22/2022]
Abstract
Aging is an inevitable risk factor for many diseases, including cardiovascular diseases, neurodegenerative diseases, cancer, and diabetes. Investigation into the molecular mechanisms involved in aging and longevity will benefit the treatment of age-dependent diseases and the development of preventative medicine for agingrelated diseases. Current evidence has revealed that FoxO3, encoding the transcription factor (FoxO)3, a key transcription factor that integrates different stimuli in the intrinsic and extrinsic pathways and is involved in cell differentiation, protein homeostasis, stress resistance and stem cell status, plays a regulatory role in longevity and in age-related diseases. However, the precise mechanisms by which the FoxO3 transcription factor modulates aging and promotes longevity have been unclear until now. Here, we provide a brief overview of the mechanisms by which FoxO3 mediates signaling in pathways involved in aging and aging-related diseases, as well as the current knowledge on the role of the FoxO3 transcription factor in the human lifespan and its clinical prospects. Ultimately, we conclude that FoxO3 signaling pathways, including upstream and downstream molecules, may be underlying therapeutic targets in aging and age-related diseases.
Collapse
Affiliation(s)
- Zao-Shang Chang
- Department of Physiology, School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, Hunan, China
| | - Zhi-Ming He
- Department of Physiology, School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, Hunan, China
| | - Jing-Bo Xia
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou 510500, Guangdong, China
| |
Collapse
|
187
|
Li S, Jakobs TC. Secreted phosphoprotein 1 slows neurodegeneration and rescues visual function in mouse models of aging and glaucoma. Cell Rep 2022; 41:111880. [PMID: 36577373 PMCID: PMC9847489 DOI: 10.1016/j.celrep.2022.111880] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/01/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
Aging causes an irreversible, cumulative decline in neuronal function. Using the visual system as a model, we show that astrocytes play a critical role in maintaining retinal ganglion cell health and that deletion of SPP1 (secreted phosphoprotein 1, or osteopontin) from astrocytes leads to increased vulnerability of ganglion cells to age, elevated intraocular pressure, and traumatic optic nerve damage. Overexpression of SPP1 slows the age-related decline in ganglion cell numbers and is highly protective of visual function in a mouse model of glaucoma. SPP1 acts by promoting phagocytosis and secretion of neurotrophic factors while inhibiting production of neurotoxic and pro-inflammatory factors. SPP1 up-regulates transcription of genes related to oxidative phosphorylation, functionally enhances mitochondrial respiration, and promotes the integrity of mitochondrial microstructure. SPP1 increases intracellular ATP concentration via up-regulation of VDAC1.
Collapse
Affiliation(s)
- Song Li
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA 02114, USA.
| | - Tatjana C Jakobs
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA 02114, USA.
| |
Collapse
|
188
|
Wu IC, Liu CS, Cheng WL, Lin TT, Chen HL, Chen PF, Wu RC, Huang CW, Hsiung CA, Hsu CC. Association of leukocyte mitochondrial DNA copy number with longitudinal C-reactive protein levels and survival in older adults: a cohort study. Immun Ageing 2022; 19:62. [PMID: 36494677 PMCID: PMC9733307 DOI: 10.1186/s12979-022-00322-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Systemic chronic inflammation occurs with age. The association of the leukocyte mitochondrial DNA copy number, a measure of mitochondrial function in aging, with the temporal profile of serum high-sensitivity C-reactive protein and mortality risk remains uncertain. The objectives of this study were to examine the association of the leukocyte mitochondrial DNA copy number with longitudinal high-sensitivity C-reactive protein levels and the association of the longitudinal high-sensitivity C-reactive protein levels with mortality risk. METHODS This prospective cohort study included 3928 adults aged ≥ 55 years without systemic inflammation in the baseline examination of the Healthy Aging Longitudinal Study in Taiwan, which started in 2009. Each participant received leukocyte mitochondrial DNA copy number measurement using a fluorescence-based quantitative polymerase chain reaction at baseline, serum high-sensitivity C-reactive protein measurements at baseline and the follow-up examination five years later, and the ascertainment of all-cause death (until November 30, 2021). The relationships among the leukocyte mitochondrial DNA copy number, longitudinal serum high-sensitivity C-reactive protein levels, and time to all-cause mortality were examined using the joint longitudinal and survival modeling analysis. RESULTS Of the 3928 participants (mean age: 69 years; 2060 [52%] were women), 837 (21%) died during follow-up. In the adjusted analysis, one standard deviation lower natural log-transformed baseline leukocyte mitochondrial DNA copy number was associated with an increase of 0.05 (95% confidence interval [CI], 0.02 to 0.08) standard deviation in serum high-sensitivity C-reactive protein in subsequent years. An increase of 1 standard deviation in instantaneous high-sensitivity C-reactive protein levels was associated with a hazard ratio (HR) for all-cause mortality of 1.22 (95% CI, 1.14 to 1.30). Similar results were obtained after further adjusting for baseline high-sensitivity C-reactive protein levels (HR [95% CI], 1.27 [1.16 to 1.38]) and after excluding those with serum high-sensitivity C-reactive protein above 10 mg/L (HR [95% CI], 1.21[1.11 to 1.31]) or 3 mg/L (HR [95% CI], 1.19 [1.06 to 1.31]) during follow-up. CONCLUSIONS A lower leukocyte mitochondrial DNA copy number was associated with persistently higher high-sensitivity C-reactive protein levels. Moreover, these higher time-varying high-sensitivity C-reactive protein levels were instantaneously associated with a higher risk of death.
Collapse
Affiliation(s)
- I-Chien Wu
- grid.59784.370000000406229172Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053 Taiwan
| | - Chin-San Liu
- grid.413814.b0000 0004 0572 7372Vascular and Genomic Center, Institute of ATP, Changhua Christian Hospital, Changhua, Taiwan ,grid.413814.b0000 0004 0572 7372Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan ,grid.254145.30000 0001 0083 6092Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan ,grid.260542.70000 0004 0532 3749Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wen-Ling Cheng
- grid.413814.b0000 0004 0572 7372Vascular and Genomic Center, Institute of ATP, Changhua Christian Hospital, Changhua, Taiwan
| | - Ta-Tsung Lin
- grid.413814.b0000 0004 0572 7372Vascular and Genomic Center, Institute of ATP, Changhua Christian Hospital, Changhua, Taiwan
| | - Hui-Ling Chen
- grid.59784.370000000406229172Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053 Taiwan
| | - Pei-Fen Chen
- grid.59784.370000000406229172Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053 Taiwan
| | - Ray-Chin Wu
- grid.59784.370000000406229172Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053 Taiwan
| | - Chen-Wei Huang
- grid.59784.370000000406229172Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053 Taiwan
| | - Chao A. Hsiung
- grid.59784.370000000406229172Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053 Taiwan
| | - Chih-Cheng Hsu
- grid.59784.370000000406229172Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053 Taiwan
| |
Collapse
|
189
|
Lv X, Gao F, Cao X. Skeletal interoception in bone homeostasis and pain. Cell Metab 2022; 34:1914-1931. [PMID: 36257317 PMCID: PMC9742337 DOI: 10.1016/j.cmet.2022.09.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023]
Abstract
Accumulating evidence indicates that interoception maintains proper physiological status and orchestrates metabolic homeostasis by regulating feeding behaviors, glucose balance, and lipid metabolism. Continuous skeletal remodeling consumes a tremendous amount of energy to provide skeletal scaffolding, support muscle movement, store vital minerals, and maintain a niche for hematopoiesis, which are processes that also contribute to overall metabolic balance. Although skeletal innervation has been described for centuries, recent work has shown that skeletal metabolism is tightly regulated by the nervous system and that skeletal interoception regulates bone homeostasis. Here, we provide a general discussion of interoception and its effects on the skeleton and whole-body metabolism. We also discuss skeletal interoception-mediated regulation in the context of pathological conditions and skeletal pain as well as future challenges to our understanding of these process and how they can be leveraged for more effective therapy.
Collapse
Affiliation(s)
- Xiao Lv
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Feng Gao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xu Cao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
190
|
Cai Y, Song W, Li J, Jing Y, Liang C, Zhang L, Zhang X, Zhang W, Liu B, An Y, Li J, Tang B, Pei S, Wu X, Liu Y, Zhuang CL, Ying Y, Dou X, Chen Y, Xiao FH, Li D, Yang R, Zhao Y, Wang Y, Wang L, Li Y, Ma S, Wang S, Song X, Ren J, Zhang L, Wang J, Zhang W, Xie Z, Qu J, Wang J, Xiao Y, Tian Y, Wang G, Hu P, Ye J, Sun Y, Mao Z, Kong QP, Liu Q, Zou W, Tian XL, Xiao ZX, Liu Y, Liu JP, Song M, Han JDJ, Liu GH. The landscape of aging. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2354-2454. [PMID: 36066811 PMCID: PMC9446657 DOI: 10.1007/s11427-022-2161-3] [Citation(s) in RCA: 190] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
Aging is characterized by a progressive deterioration of physiological integrity, leading to impaired functional ability and ultimately increased susceptibility to death. It is a major risk factor for chronic human diseases, including cardiovascular disease, diabetes, neurological degeneration, and cancer. Therefore, the growing emphasis on "healthy aging" raises a series of important questions in life and social sciences. In recent years, there has been unprecedented progress in aging research, particularly the discovery that the rate of aging is at least partly controlled by evolutionarily conserved genetic pathways and biological processes. In an attempt to bring full-fledged understanding to both the aging process and age-associated diseases, we review the descriptive, conceptual, and interventive aspects of the landscape of aging composed of a number of layers at the cellular, tissue, organ, organ system, and organismal levels.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Wei Song
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Jing
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuqian Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Liyuan Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Xia Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenhui Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Beibei Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Yongpan An
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Baixue Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xueying Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuxuan Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Cheng-Le Zhuang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Xuefeng Dou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fu-Hui Xiao
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- The Fifth People's Hospital of Chongqing, Chongqing, 400062, China.
| | - Xiaoyuan Song
- MOE Key Laboratory of Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liang Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jun Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ye Tian
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Gelin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Ping Hu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, 98195, USA.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Qiang Liu
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| | - Zhi-Xiong Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| | - Yong Liu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China.
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China.
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, Victoria, 3181, Australia.
- Hudson Institute of Medical Research, and Monash University Department of Molecular and Translational Science, Clayton, Victoria, 3168, Australia.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
191
|
Lu S, Li C, Jin X, Zhu L, Shen J, Bai M, Li Y, Xu E. Baicalin improves the energy levels in the prefrontal cortex of mice exposed to chronic unpredictable mild stress. Heliyon 2022; 8:e12083. [DOI: 10.1016/j.heliyon.2022.e12083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/11/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
|
192
|
Hu Y, Cao K, Wang F, Wu W, Mai W, Qiu L, Luo Y, Ge WP, Sun B, Shi L, Zhu J, Zhang J, Wu Z, Xie Y, Duan S, Gao Z. Dual roles of hexokinase 2 in shaping microglial function by gating glycolytic flux and mitochondrial activity. Nat Metab 2022; 4:1756-1774. [PMID: 36536134 DOI: 10.1038/s42255-022-00707-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/04/2022] [Indexed: 12/24/2022]
Abstract
Microglia continuously survey the brain parenchyma and actively shift status following stimulation. These processes demand a unique bioenergetic programme; however, little is known about the metabolic determinants in microglia. By mining large datasets and generating transgenic tools, here we show that hexokinase 2 (HK2), the most active isozyme associated with mitochondrial membrane, is selectively expressed in microglia in the brain. Genetic ablation of HK2 reduced microglial glycolytic flux and energy production, suppressed microglial repopulation, and attenuated microglial surveillance and damage-triggered migration in male mice. HK2 elevation is prominent in immune-challenged or disease-associated microglia. In ischaemic stroke models, however, HK2 deletion promoted neuroinflammation and potentiated cerebral damages. The enhanced inflammatory responses after HK2 ablation in microglia are associated with aberrant mitochondrial function and reactive oxygen species accumulation. Our study demonstrates that HK2 gates both glycolytic flux and mitochondrial activity to shape microglial functions, changes of which contribute to metabolic abnormalities and maladaptive inflammation in brain diseases.
Collapse
Affiliation(s)
- Yaling Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Kelei Cao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Fang Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weiying Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Liyao Qiu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yuxiang Luo
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing, Beijing, China
| | - Binggui Sun
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Ligen Shi
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junming Zhu
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiying Wu
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yicheng Xie
- The Children's Hospital, Zhejiang, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
193
|
Xu L, Li L, Pan C, Song J, Zhang C, Wu X, Hu F, Liu X, Zhang Z, Zhang Z. Erythropoietin signaling in peripheral macrophages is required for systemic β-amyloid clearance. EMBO J 2022; 41:e111038. [PMID: 36215698 PMCID: PMC9670197 DOI: 10.15252/embj.2022111038] [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: 02/28/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 01/13/2023] Open
Abstract
Impaired clearance of beta-amyloid (Aβ) is a primary cause of sporadic Alzheimer's disease (AD). Aβ clearance in the periphery contributes to reducing brain Aβ levels and preventing Alzheimer's disease pathogenesis. We show here that erythropoietin (EPO) increases phagocytic activity, levels of Aβ-degrading enzymes, and Aβ clearance in peripheral macrophages via PPARγ. Erythropoietin is also shown to suppress Aβ-induced inflammatory responses. Deletion of EPO receptor in peripheral macrophages leads to increased peripheral and brain Aβ levels and exacerbates Alzheimer's-associated brain pathologies and behavioral deficits in AD-model mice. Moreover, erythropoietin signaling is impaired in peripheral macrophages of old AD-model mice. Exogenous erythropoietin normalizes impaired EPO signaling and dysregulated functions of peripheral macrophages in old AD-model mice, promotes systemic Aβ clearance, and alleviates disease progression. Erythropoietin treatment may represent a potential therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Lu Xu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
- Department of Neurology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Lei Li
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Cai‐Long Pan
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
| | - Jing‐Jing Song
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Chen‐Yang Zhang
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Xiang‐Hui Wu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Fan Hu
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Xue Liu
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Zhiren Zhang
- Institute of ImmunologyArmy Medical UniversityChongqingChina
| | - Zhi‐Yuan Zhang
- School of Basic Medical SciencesNanjing Medical UniversityNanjingChina
- Key Laboratory of Antibody Technique of Ministry of HealthNanjing Medical UniversityNanjingChina
- Department of Neurology, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
- Key Laboratory of Human Functional Genomics of Jiangsu ProvinceNanjing Medical UniversityNanjingChina
| |
Collapse
|
194
|
Guan X, Liu Y, Xin W, Qin S, Gong S, Xiao T, Zhang D, Li Y, Xiong J, Yang K, He T, Zhao J, Huang Y. Activation of EP4 alleviates AKI-to-CKD transition through inducing CPT2-mediated lipophagy in renal macrophages. Front Pharmacol 2022; 13:1030800. [PMID: 36467025 PMCID: PMC9709464 DOI: 10.3389/fphar.2022.1030800] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/04/2022] [Indexed: 02/01/2025] Open
Abstract
Acute kidney injury (AKI) is a common clinical syndrome with complex pathogenesis, characterized by a rapid decline in kidney function in the short term. Worse still, the incomplete recovery from AKI increases the risk of progression to chronic kidney disease (CKD). However, the pathogenesis and underlying mechanism remain largely unknown. Macrophages play an important role during kidney injury and tissue repair, but its role in AKI-to-CKD transition remains elusive. Herein, single nucleus RNA sequencing (snRNA-Seq) and flow cytometry validations showed that E-type prostaglandin receptor 4 (EP4) was selectively activated in renal macrophages, rather than proximal tubules, in ischemia-reperfusion injury (IRI)-induced AKI-to-CKD transition mouse model. EP4 inhibition aggravated AKI-to-CKD transition, while EP4 activation impeded the progression of AKI to CKD though regulating macrophage polarization. Mechanistically, network pharmacological analysis and subsequent experimental verifications revealed that the activated EP4 inhibited macrophage polarization through inducing Carnitine palmitoyltransferase 2 (CPT2)-mediated lipophagy in macrophages. Further, CPT2 inhibition abrogated the protective effect of EP4 on AKI-to-CKD transition. Taken together, our findings demonstrate that EP4-CPT2 signaling-mediated lipophagy in macrophages plays a pivotal role in the transition of AKI to CKD and targeting EP4-CPT2 axis could serve as a promising therapeutic approach for retarding AKI and its progression to CKD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jinghong Zhao
- The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Department of Nephrology, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yinghui Huang
- The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Department of Nephrology, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
195
|
Chen J, Deng JC, Zemans RL, Bahmed K, Kosmider B, Zhang M, Peters-Golden M, Goldstein DR. Age-induced prostaglandin E 2 impairs mitochondrial fitness and increases mortality to influenza infection. Nat Commun 2022; 13:6759. [PMID: 36351902 PMCID: PMC9643978 DOI: 10.1038/s41467-022-34593-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 10/31/2022] [Indexed: 11/10/2022] Open
Abstract
Aging impairs the immune responses to influenza A virus (IAV), resulting in increased mortality to IAV infections in older adults. However, the factors within the aged lung that compromise host defense to IAV remain unknown. Using a murine model and human samples, we identified prostaglandin E2 (PGE2), as such a factor. Senescent type II alveolar epithelial cells (AECs) are overproducers of PGE2 within the aged lung. PGE2 impairs the proliferation of alveolar macrophages (AMs), critical cells for defense against respiratory pathogens, via reduction of oxidative phosphorylation and mitophagy. Importantly, blockade of the PGE2 receptor EP2 in aged mice improves AM mitochondrial function, increases AM numbers and enhances survival to IAV infection. In conclusion, our study reveals a key mechanism that compromises host defense to IAV, and possibly other respiratory infections, with aging and suggests potential new therapeutic or preventative avenues to protect against viral respiratory disease in older adults.
Collapse
Affiliation(s)
- Judy Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jane C Deng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rachel L Zemans
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Karim Bahmed
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA, 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, 19140, USA
| | - Beata Kosmider
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA, 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, 19140, USA
| | - Min Zhang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Marc Peters-Golden
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daniel R Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Program in Immunology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
196
|
Schwartz M, Abellanas MA, Tsitsou-Kampeli A, Suzzi S. The brain-immune ecosystem: Implications for immunotherapy in defeating neurodegenerative diseases. Neuron 2022; 110:3421-3424. [PMID: 36150394 DOI: 10.1016/j.neuron.2022.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 12/15/2022]
Abstract
Recent functional and anatomical discoveries of brain-immune relationships have overturned previous beliefs regarding the brain's immune privilege. Here, we propose that the brain and immune cells at its borders operate as an "ecosystem" to support the brain's robustness and resilience. Modulation of this ecosystem can be harnessed in the clinic.
Collapse
Affiliation(s)
- Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| | - Miguel A Abellanas
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Stefano Suzzi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
197
|
Tremblay MÈ, Almsherqi ZA, Deng Y. Plasmalogens and platelet-activating factor roles in chronic inflammatory diseases. Biofactors 2022; 48:1203-1216. [PMID: 36370412 DOI: 10.1002/biof.1916] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022]
Abstract
Fatty acids and phospholipid molecules are essential for determining the structure and function of cell membranes, and they hence participate in many biological processes. Platelet activating factor (PAF) and its precursor plasmalogen, which represent two subclasses of ether phospholipids, have attracted increasing research attention recently due to their association with multiple chronic inflammatory, neurodegenerative, and metabolic disorders. These pathophysiological conditions commonly involve inflammatory processes linked to an excess presence of PAF and/or decreased levels of plasmalogens. However, the molecular mechanisms underlying the roles of plasmalogens in inflammation have remained largely elusive. While anti-inflammatory responses most likely involve the plasmalogen signal pathway; pro-inflammatory responses recruit arachidonic acid, a precursor of pro-inflammatory lipid mediators which is released from membrane phospholipids, notably derived from the hydrolysis of plasmalogens. Plasmalogens per se are vital membrane phospholipids in humans. Changes in their homeostatic levels may alter cell membrane properties, thus affecting key signaling pathways that mediate inflammatory cascades and immune responses. The plasmalogen analogs of PAF are also potentially important, considering that anti-PAF activity has strong anti-inflammatory effects. Plasmalogen replacement therapy was further identified as a promising anti-inflammatory strategy allowing for the relief of pathological hallmarks in patients affected by chronic diseases with an inflammatory component. The aim of this Short Review is to highlight the emerging roles and implications of plasmalogens in chronic inflammatory disorders, along with the promising outcomes of plasmalogen replacement therapy for the treatment of various PAF-related chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, Canada
- Department of Molecular Medicine, Université de Laval, Québec City, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
| | - Zakaria A Almsherqi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| |
Collapse
|
198
|
Peralta Ramos JM, Kviatcovsky D, Schwartz M. Targeting the immune system towards novel therapeutic avenues to fight brain aging and neurodegeneration. Eur J Neurosci 2022; 56:5413-5427. [PMID: 35075702 DOI: 10.1111/ejn.15609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
The incidence of age-related dementia is growing with increased longevity, yet there are currently no disease-modifying therapies for these devastating disorders. Studies over the last several years have led to an evolving awareness of the role of the immune system in supporting brain maintenance and repair, displaying a diverse repertoire of functions while orchestrating the crosstalk between the periphery and the brain. Here, we provide insights into the current understanding of therapeutic targets that could be adopted to modulate immune cell fate, either systemically or locally, to defeat brain aging and neurodegeneration.
Collapse
Affiliation(s)
| | - Denise Kviatcovsky
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
199
|
Rossi E, Mutti L, Morrione A, Giordano A. Neuro-Immune Interactions in Severe COVID-19 Infection. Pathogens 2022; 11:1256. [PMID: 36365007 PMCID: PMC9699641 DOI: 10.3390/pathogens11111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a new coronavirus that has affected the world since 2019. Interstitial pneumonia is the most common clinical presentation, but additional symptoms have been reported, including neurological manifestations. Severe forms of infection, especially in elderly patients, present as an excessive inflammatory response called "cytokine storm", which can lead to acute respiratory distress syndrome (ARDS), multiorgan failure and death. Little is known about the relationship between symptoms and clinical outcomes or the characteristics of virus-host interactions. The aim of this narrative review is to highlight possible links between neurological involvement and respiratory damage mediated by pathological inflammatory pathways in SARS-CoV-2 infection. We will focus on neuro-immune interactions and age-related immunity decline and discuss some pathological mechanisms that contribute to negative outcomes in COVID-19 patients. Furthermore, we will describe available therapeutic strategies and their effects on COVID-19 neurological symptoms.
Collapse
Affiliation(s)
- Elena Rossi
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Italian Group for Research and Therapy for Mesothelioma (GIMe), 27058 Voghera, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
200
|
Kim J, Kang S, Hong H, Joo M, Kang H. A Non-Randomized Combined Program of Walking and Low-Load Resistance Exercise Improves Cognitive Function and Cardiometabolic Risk Factors in Community-Dwelling Elderly Women. Healthcare (Basel) 2022; 10:2106. [PMID: 36292553 PMCID: PMC9602658 DOI: 10.3390/healthcare10102106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/04/2022] Open
Abstract
Background: This study examines whether changes in cardiometabolic risk factors, functional fitness, and depressive symptoms following a six-month exercise intervention were associated with cognitive function in Korean women aged 65 years and older. Methods: A non-randomized study design was used to compare post-intervention changes in measured variables between control (n = 30) and exercise (n = 30) groups. The exercise intervention consisted of three days of low-load resistance exercise and two days of walking. Cognitive function and depressive symptoms were assessed with the Korean version of the Mini−Mental State Examination and the Korean version of the Geriatric Depression Scale, respectively. Functional fitness was measured using a senior fitness test battery. Results: The exercise group showed a significant improvement in cognitive function (p < 0.001) in conjunction with significant decreases in blood glucose (p = 0.052), triglycerides (p = 0.011), insulin (p = 0.002), tumor necrosis factor-α (p = 0.043), and depressive symptoms (p = 0.006) and an increase in interleukin-10 (p = 0.037), compared with the control group. Multivariate stepwise regression showed that changes in depressive symptoms (p < 0.001), insulin resistance (p < 0.001), and upper body muscle strength (p = 0.003) were positively associated with cognitive function. Conclusion: A six-month exercise intervention consisting of walking and low-load/high-repetition elastic band resistance exercise has the potential to improve cognitive function, as well as physical function and cardiometabolic risk factors, and to decrease depressive symptoms in older women.
Collapse
Affiliation(s)
| | | | | | | | - Hyunsik Kang
- College of Sport Science, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|