201
|
Monzón-Sandoval J, Burlacu E, Agarwal D, Handel AE, Wei L, Davis J, Cowley SA, Cader MZ, Webber C. Lipopolysaccharide distinctively alters human microglia transcriptomes to resemble microglia from Alzheimer's disease mouse models. Dis Model Mech 2022; 15:277958. [PMID: 36254682 PMCID: PMC9612871 DOI: 10.1242/dmm.049349] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 08/30/2022] [Indexed: 01/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, and risk-influencing genetics implicates microglia and neuroimmunity in the pathogenesis of AD. Induced pluripotent stem cell (iPSC)-derived microglia (iPSC-microglia) are increasingly used as a model of AD, but the relevance of historical immune stimuli to model AD is unclear. We performed a detailed cross-comparison over time on the effects of combinatory stimulation of iPSC-microglia, and in particular their relevance to AD. We used single-cell RNA sequencing to measure the transcriptional response of iPSC-microglia after 24 h and 48 h of stimulation with prostaglandin E2 (PGE2) or lipopolysaccharide (LPS)+interferon gamma (IFN-γ), either alone or in combination with ATPγS. We observed a shared core transcriptional response of iPSC-microglia to ATPγS and to LPS+IFN-γ, suggestive of a convergent mechanism of action. Across all conditions, we observed a significant overlap, although directional inconsistency to genes that change their expression levels in human microglia from AD patients. Using a data-led approach, we identify a common axis of transcriptomic change across AD genetic mouse models of microglia and show that only LPS provokes a transcriptional response along this axis in mouse microglia and LPS+IFN-γ in human iPSC-microglia. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Elena Burlacu
- John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
| | - Devika Agarwal
- Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK.,Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Adam E Handel
- John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK
| | - Liting Wei
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - John Davis
- Nuffield Department of Medicine Research Building, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Sally A Cowley
- Translational Molecular Neuroscience Group, New Biochemistry Building, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3QU, UK
| | - M Zameel Cader
- John Radcliffe Hospital, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DS, UK.,James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Caleb Webber
- UK Dementia Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
202
|
St-Pierre MK, Carrier M, González Ibáñez F, Šimončičová E, Wallman MJ, Vallières L, Parent M, Tremblay MÈ. Ultrastructural characterization of dark microglia during aging in a mouse model of Alzheimer's disease pathology and in human post-mortem brain samples. J Neuroinflammation 2022; 19:235. [PMID: 36167544 PMCID: PMC9513936 DOI: 10.1186/s12974-022-02595-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/12/2022] [Indexed: 11/10/2022] Open
Abstract
A diverse heterogeneity of microglial cells was previously described in Alzheimer's disease (AD) pathology, including dark microglia, a state characterized by ultrastructural markers of cellular stress. To provide novel insights into the roles of dark microglia during aging in the context of AD pathology, we performed a quantitative density and ultrastructural analysis of these cells using high-throughput scanning electron microscopy in the ventral hippocampus CA1 stratum lacunosum-moleculare of 20-month-old APP-PS1 vs C57BL/6J male mice. The density of dark microglia was significantly higher in APP-PS1 vs C57BL/6J mice, with these cells accounting for nearly half of all microglia observed near amyloid-beta (Aβ) plaques. This dark microglial state interacted more with dystrophic neurites compared to other APP-PS1 microglia and possessed glycogen granules, associated with a metabolic shift toward glycolysis, which provides the first ultrastructural evidence of their presence in microglia. Dark microglia were further observed in aging human post-mortem brain samples showing similar ultrastructural features as in mouse. Overall, our results provide a quantitative ultrastructural characterization of a microglial state associated with cellular stress (i.e., dark microglia) that is primarily restricted near Aβ plaques and dystrophic neurites. The presence of this microglial state in the aging human post-mortem brain is further revealed.
Collapse
Affiliation(s)
- Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Fernando González Ibáñez
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Marie-Josée Wallman
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Quebec, QC, Canada.,CERVO Brain Research Center, Quebec, QC, Canada
| | - Luc Vallières
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
| | - Martin Parent
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Quebec, QC, Canada.,CERVO Brain Research Center, Quebec, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada. .,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada. .,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada. .,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada. .,Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
203
|
Abstract
To maintain energy supply to the brain, a direct energy source called adenosine triphosphate (ATP) is produced by oxidative phosphorylation and aerobic glycolysis of glucose in the mitochondria and cytoplasm. Brain glucose metabolism is reduced in many neurodegenerative diseases, including Alzheimer's disease (AD), where it appears presymptomatically in a progressive and region-specific manner. Following dysregulation of energy metabolism in AD, many cellular repair/regenerative processes are activated to conserve the energy required for cell viability. Glucose metabolism plays an important role in the pathology of AD and is closely associated with the tricarboxylic acid cycle, type 2 diabetes mellitus, and insulin resistance. The glucose intake in neurons is from endothelial cells, astrocytes, and microglia. Damage to neurocentric glucose also damages the energy transport systems in AD. Gut microbiota is necessary to modulate bidirectional communication between the gastrointestinal tract and brain. Gut microbiota may influence the process of AD by regulating the immune system and maintaining the integrity of the intestinal barrier. Furthermore, some therapeutic strategies have shown promising therapeutic effects in the treatment of AD at different stages, including the use of antidiabetic drugs, rescuing mitochondrial dysfunction, and epigenetic and dietary intervention. This review discusses the underlying mechanisms of alterations in energy metabolism in AD and provides potential therapeutic strategies in the treatment of AD.
Collapse
|
204
|
Hua Y, Chen S, Xiong X, Lin C, Li D, Tu P. Risk factors for postoperative delirium in elderly urological patients: A meta-analysis. Medicine (Baltimore) 2022; 101:e30696. [PMID: 36197185 PMCID: PMC9509152 DOI: 10.1097/md.0000000000030696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Avoiding postoperative delirium (POD) can have a significant detrimental effect on the rehabilitation and prognosis of elderly urological patients. It is necessary to explore the risk factors associated with POD in elderly urology to provide a basis for clinical recognition of delirium. METHODS For relevant studies, we comprehensively searched Embase, MEDLINE, Ovid, PubMed, Scopus, The Cochrane Library, and Web of Science. The search deadline was September 2021. RESULTS We identified 2046 studies, 8 of which were included in the ultimate analysis. A total of 8 articles, including 356 cases in the delirium group and 1813 cases in the non-delirium group, were included in the relevant literature. The 2 groups mentioned above differed significantly in the following factors: history of delirium (odds ratio [OR] = 6.98, 95% confidence interval [CI]: 1.63-29.86, P = .009); Preoperative use of psychotropic drugs (OR = 1.97, 95% CI: 1.11-3.52, P = .02); age (OR = 3.10, 95% CI: 2.08-4.12, P < .0001). The meta-analysis demonstrated that smoking, alcohol consumption, gender (male), mode of anesthesia (general anesthesia) and being unmarried did not have a significant effect on POD in elderly urological patients. CONCLUSION The risk factors for POD in elderly urological patients include history of delirium, preoperative use of psychotropic drugs, and age. The present study provides guidance for taking targeted preventive measures to reduce risks.
Collapse
Affiliation(s)
- Yaqi Hua
- School of Nursing, Nanchang University, Nanchang, China
| | - Shoulin Chen
- Department of Anesthesia, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoyun Xiong
- Department of Nursing, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chuyang Lin
- Department of Clinical Trial Research Center, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dongying Li
- Department of Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ping Tu
- Department of Post-Anesthesia Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Ping Tu, Department of Post-Anesthesia Care Unit, The Second Affiliated Hospital of Nanchang University, 1 Min DE Road, Nanchang 330006, China (e-mail: )
| |
Collapse
|
205
|
Afridi R, Rahman MH, Suk K. Implications of glial metabolic dysregulation in the pathophysiology of neurodegenerative diseases. Neurobiol Dis 2022; 174:105874. [PMID: 36154877 DOI: 10.1016/j.nbd.2022.105874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 08/28/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Glial cells are the most abundant cells of the brain, outnumbering neurons. These multifunctional cells are crucial for maintaining brain homeostasis by providing trophic and nutritional support to neurons, sculpting synapses, and providing an immune defense. Glia are highly plastic and undergo both structural and functional alterations in response to changes in the brain microenvironment. Glial phenotypes are intimately regulated by underlying metabolic machinery, which dictates the effector functions of these cells. Altered brain energy metabolism and chronic neuroinflammation are common features of several neurodegenerative diseases. Microglia and astrocytes are the major glial cells fueling the ongoing neuroinflammatory process, exacerbating neurodegeneration. Distinct metabolic perturbations in microglia and astrocytes, including altered carbohydrate, lipid, and amino acid metabolism have been documented in neurodegenerative diseases. These disturbances aggravate the neurodegenerative process by potentiating the inflammatory activation of glial cells. This review covers the recent advances in the molecular aspects of glial metabolic changes in the pathophysiology of neurodegenerative diseases. Finally, we discuss studies exploiting glial metabolism as a potential therapeutic avenue in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
206
|
Zhu R, Zeng S, Li N, Fu N, Wang Y, Miao M, Yang Y, Sun M, Zhang J. Sevoflurane exposure induces neurotoxicity by regulating mitochondrial function of microglia due to NAD insufficiency. Front Cell Neurosci 2022; 16:914957. [PMID: 36212689 PMCID: PMC9532507 DOI: 10.3389/fncel.2022.914957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Developmental neurons received with sevoflurane, the commonly used inhalational anesthetic agent in clinical surgery, several times tend to be destroyed. Microglia, the resident immune cells of the central nervous system (CNS), are activated after sevoflurane exposure, accompanied by releasing proinflammatory cytokines that damage developing neurons. The sevoflurane-induced neurotoxicity could be attributed to activated microglia presenting proinflammatory and anti-inflammatory functions. Proinflammatory microglia release cytokines to impair the CNS, while anti-inflammatory microglia engulf damaged neurons to maintain CNS homeostasis. Sevoflurane exposure promotes the secretion of proinflammatory cytokines by microglia, inhibiting the microglial phagocytic function. Microglia with poor phagocytic function cannot engulf damaged neurons, leading to the accumulation of damaged neurons. The mechanism underlying poor phagocytic function may be attributed to mitochondrial dysfunction of microglia induced by sevoflurane exposure, in which affected mitochondria cannot generate adequate ATP and NAD to satisfy the energy demand. We discovered that sevoflurane treatment impaired the mitochondrial metabolism of microglia, which resulted in NAD deficiency and couldn’t produce sufficient energy to clear damaged neurons to maintain CNS development. Our findings provide an explanation of a new mechanism underlying sevoflurane-induced neurotoxicity.
Collapse
|
207
|
Xia A, Thai A, Cao Z, Chen X, Chen J, Bacacao B, Bekale LA, Schiel V, Bollyky PL, Maria PLS. Chronic suppurative otitis media causes macrophage-associated sensorineural hearing loss. J Neuroinflammation 2022; 19:224. [PMID: 36096817 PMCID: PMC9465898 DOI: 10.1186/s12974-022-02585-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Chronic suppurative otitis media (CSOM) is the most common cause of permanent hearing loss in children in the developing world. A large component of the permanent hearing loss is sensory in nature and our understanding of the mechanism of this has so far been limited to post-mortem human specimens or acute infection models that are not representative of human CSOM. In this report, we assess cochlear injury in a validated Pseudomonas aeruginosa (PA) CSOM mouse model. Methods We generated persisters (PCs) and inoculated them into the mouse middle ear cavity. We tracked infection with IVIS and detected PA using RT-PCR. We assessed cochlear damage and innate immunity by Immunohistochemistry. Finally, we evaluated cytokines with multiplex assay and quantitative real-time PCR. Results We observed outer hair cell (OHC) loss predominantly in the basal turn of the cochlear at 14 days after bacterial inoculation. Macrophages, not neutrophils are the major immune cells in the cochlea in CSOM displaying increased numbers and a distribution correlated with the observed cochlear injury. The progression of the morphological changes suggests a transition from monocytes into tissue macrophages following infection. We also show that PA do not enter the cochlea and live bacteria are required for cochlear injury. We characterized cytokine activity in the CSOM cochlea. Conclusions Taken together, this data shows a critical role for macrophages in CSOM-mediated sensorineural hearing loss (SNHL). Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02585-w.
Collapse
|
208
|
Ye F, Funk Q, Rockers E, Shulman JM, Masdeu JC, Pascual B. In Alzheimer-prone brain regions, metabolism and risk-gene expression are strongly correlated. Brain Commun 2022; 4:fcac216. [PMID: 36092303 PMCID: PMC9453434 DOI: 10.1093/braincomms/fcac216] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/20/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Neuroimaging in the preclinical phase of Alzheimer’s disease provides information crucial to early intervention, particularly in people with a high genetic risk. Metabolic network modularity, recently applied to the study of dementia, is increased in Alzheimer’s disease patients compared with controls, but network modularity in cognitively unimpaired elderly with various risks of developing Alzheimer’s disease needs to be determined. Based on their 5-year cognitive progression, we stratified 117 cognitively normal participants (78.3 ± 4.0 years of age, 52 women) into three age-matched groups, each with a different level of risk for Alzheimer’s disease. From their fluorodeoxyglucose PET we constructed metabolic networks, evaluated their modular structures using the Louvain algorithm, and compared them between risk groups. As the risk for Alzheimer’s disease increased, the metabolic connections among brain regions weakened and became more modular, indicating network fragmentation and functional impairment of the brain. We then set out to determine the correlation between regional brain metabolism, particularly in the modules derived from the previous analysis, and the regional expression of Alzheimer-risk genes in the brain, obtained from the Allen Human Brain Atlas. In all risk groups of this elderly population, the regional brain expression of most Alzheimer-risk genes showed a strong correlation with brain metabolism, particularly in the module that corresponded to regions of the brain that are affected earliest and most severely in Alzheimer’s disease. Among the genes, APOE and CD33 showed the strongest negative correlation and SORL1 showed the strongest positive correlation with brain metabolism. The Pearson correlation coefficients remained significant when contrasted against a null-hypothesis distribution of correlation coefficients across the whole transcriptome of 20 736 genes (SORL1: P = 0.0130; CD33, P = 0.0136; APOE: P = 0.0093). The strong regional correlation between Alzheimer-related gene expression in the brain and brain metabolism in older adults highlights the role of brain metabolism in the genesis of dementia.
Collapse
Affiliation(s)
- Fengdan Ye
- Department of Physics and Astronomy, Rice University , Houston, TX 77005 , USA
- Center for Theoretical Biological Physics, Rice University , Houston, TX 77005 , USA
- Nantz National Alzheimer Center, Houston Methodist Neurological and Research Institute, Houston Methodist Hospital, Weill Cornell Medicine , Houston, TX 77030 , USA
| | - Quentin Funk
- Nantz National Alzheimer Center, Houston Methodist Neurological and Research Institute, Houston Methodist Hospital, Weill Cornell Medicine , Houston, TX 77030 , USA
| | - Elijah Rockers
- Nantz National Alzheimer Center, Houston Methodist Neurological and Research Institute, Houston Methodist Hospital, Weill Cornell Medicine , Houston, TX 77030 , USA
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine , Houston, TX 77030 , USA
- Department of Neuroscience, Baylor College of Medicine , Houston, TX 77030 , USA
- Department of Molecular and Human Genetics, Baylor College of Medicine , Houston, TX 77030 , USA
- Center for Alzheimer’s and Neurodegenerative Diseases, Baylor College of Medicine , Houston, TX 77030 , USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital , Houston, TX 77030 , USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center, Houston Methodist Neurological and Research Institute, Houston Methodist Hospital, Weill Cornell Medicine , Houston, TX 77030 , USA
| | - Belen Pascual
- Nantz National Alzheimer Center, Houston Methodist Neurological and Research Institute, Houston Methodist Hospital, Weill Cornell Medicine , Houston, TX 77030 , USA
| | | |
Collapse
|
209
|
De Santi C, Nally FK, Afzal R, Duffy CP, Fitzsimons S, Annett SL, Robson T, Dowling JK, Cryan SA, McCoy CE. Enhancing arginase 2 expression using target site blockers as a strategy to modulate macrophage phenotype. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:643-655. [PMID: 36090747 PMCID: PMC9424864 DOI: 10.1016/j.omtn.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022]
Abstract
Macrophages are plastic cells playing a crucial role in innate immunity. While fundamental in responding to infections, when persistently maintained in a pro-inflammatory state they can initiate and sustain inflammatory diseases. Therefore, a strategy that reprograms pro-inflammatory macrophages toward an anti-inflammatory phenotype could hold therapeutic potential in that context. We have recently shown that arginase 2 (Arg2), a mitochondrial enzyme involved in arginine metabolism, promotes the resolution of inflammation in macrophages and it is targeted by miR-155. Here, we designed and tested a target site blocker (TSB) that specifically interferes and blocks the interaction between miR-155 and Arg2 mRNA, leading to Arg2 increased expression and activity. In bone marrow-derived macrophages transfected with Arg2 TSB (in the presence or absence of the pro-inflammatory stimulus LPS), we observed an overall shift of the polarization status of macrophages toward an anti-inflammatory phenotype, as shown by significant changes in surface markers (CD80 and CD71), metabolic parameters (mitochondrial oxidative phosphorylation) and cytokines secretion (IL-1β, IL-6, and TNF). Moreover, in an in vivo model of LPS-induced acute inflammation, intraperitoneal administration of Arg2 TSB led to an overall decrease in systemic levels of pro-inflammatory cytokines. Overall, this proof-of-concept strategy represent a promising approach to modulating macrophage phenotype.
Collapse
Affiliation(s)
- Chiara De Santi
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Corresponding author Chiara De Santi, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland.
| | - Frances K. Nally
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Remsha Afzal
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Conor P. Duffy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Stephen Fitzsimons
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Stephanie L. Annett
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Jennifer K. Dowling
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Claire E. McCoy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| |
Collapse
|
210
|
Ao Z, Song S, Tian C, Cai H, Li X, Miao Y, Wu Z, Krzesniak J, Ning B, Gu M, Lee LP, Guo F. Understanding Immune-Driven Brain Aging by Human Brain Organoid Microphysiological Analysis Platform. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200475. [PMID: 35908805 PMCID: PMC9507385 DOI: 10.1002/advs.202200475] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/17/2022] [Indexed: 05/09/2023]
Abstract
The aging of the immune system drives systemic aging and the pathogenesis of age-related diseases. However, a significant knowledge gap remains in understanding immune-driven aging, especially in brain aging, due to the limited current in vitro models of neuroimmune interaction. Here, the authors report the development of a human brain organoid microphysiological analysis platform (MAP) to discover the dynamic process of immune-driven brain aging. The organoid MAP is created by 3D printing that confines organoid growth and facilitates cell and nutrition perfusion, promoting organoid maturation and their committment to forebrain identity. Dynamic rocking flow is incorporated into the platform that allows to perfuse primary monocytes from young (20 to 30-year-old) and aged (>60-year-old) donors and culture human cortical organoids to model neuroimmune interaction. The authors find that the aged monocytes increase infiltration and promote the expression of aging-related markers (e.g., higher expression of p16) within the human cortical organoids, indicating that aged monocytes may drive brain aging. The authors believe that the organoid MAP may provide promising solutions for basic research and translational applications in aging, neural immunological diseases, autoimmune disorders, and cancer.
Collapse
Affiliation(s)
- Zheng Ao
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Sunghwa Song
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Chunhui Tian
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Hongwei Cai
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Xiang Li
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Yifei Miao
- Center for Stem Cell and Organoid Medicine (CuSTOM)Division of Pulmonary BiologyDivision of Developmental BiologyCincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
- University of Cincinnati School of MedicineCincinnatiOH45229USA
| | - Zhuhao Wu
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Jonathan Krzesniak
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Bo Ning
- Center for Cellular and Molecular DiagnosticsDepartment of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLA70112USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM)Division of Pulmonary BiologyDivision of Developmental BiologyCincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
- University of Cincinnati School of MedicineCincinnatiOH45229USA
| | - Luke P. Lee
- Harvard Institute of MedicineHarvard Medical SchoolHarvard UniversityBrigham and Women's HospitalBostonMA02115USA
- Department of BioengineeringDepartment of Electrical Engineering and Computer ScienceUniversity of California at BerkeleyBerkeleyCA94720USA
- Department of BiophysicsInstitute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi‐do16419South Korea
| | - Feng Guo
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| |
Collapse
|
211
|
Lonnemann N, Hosseini S, Ohm M, Geffers R, Hiller K, Dinarello CA, Korte M. IL-37 expression reduces acute and chronic neuroinflammation and rescues cognitive impairment in an Alzheimer's disease mouse model. eLife 2022; 11:75889. [PMID: 36040311 PMCID: PMC9481244 DOI: 10.7554/elife.75889] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The anti-inflammatory cytokine interleukin-37 (IL-37) belongs to the IL-1 family but is not expressed in mice. We used a human IL-37 (hIL-37tg) expressing mouse, which has been subjected to various models of local and systemic inflammation as well as immunological challenges. Previous studies reveal an immunomodulatory role of IL-37, which can be characterized as an important suppressor of innate immunity. Here, we examined the functions of IL-37 in the central nervous system and explored the effects of IL-37 on neuronal architecture and function, microglial phenotype, cytokine production and behavior after inflammatory challenge by intraperitoneal LPS-injection. In wild-type mice, decreased spine density, activated microglial phenotype and impaired long-term potentiation (LTP) were observed after LPS injection, whereas hIL-37tg mice showed no impairment. In addition, we crossed the hIL-37tg mouse with an animal model of Alzheimer’s disease (APP/PS1) to investigate the anti-inflammatory properties of IL-37 under chronic neuroinflammatory conditions. Our results show that expression of IL-37 is able to limit inflammation in the brain after acute inflammatory events and prevent loss of cognitive abilities in a mouse model of AD.
Collapse
Affiliation(s)
- Niklas Lonnemann
- Department of Cellular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Melanie Ohm
- Department of Cellular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Karsten Hiller
- Braunschweig Integrated Centre of Systems Biology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Health, Aurora, United States
| | - Martin Korte
- Department of Cellular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
212
|
Tyler SEB, Tyler LDK. Therapeutic roles of plants for 15 hypothesised causal bases of Alzheimer's disease. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:34. [PMID: 35996065 PMCID: PMC9395556 DOI: 10.1007/s13659-022-00354-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 05/26/2023]
Abstract
Alzheimer's disease (AD) is progressive and ultimately fatal, with current drugs failing to reverse and cure it. This study aimed to find plant species which may provide therapeutic bioactivities targeted to causal agents proposed to be driving AD. A novel toolkit methodology was employed, whereby clinical symptoms were translated into categories recognized in ethnomedicine. These categories were applied to find plant species with therapeutic effects, mined from ethnomedical surveys. Survey locations were mapped to assess how this data is at risk. Bioactivities were found of therapeutic relevance to 15 hypothesised causal bases for AD. 107 species with an ethnological report of memory improvement demonstrated therapeutic activity for all these 15 causal bases. The majority of the surveys were found to reside within biodiversity hotspots (centres of high biodiversity under threat), with loss of traditional knowledge the most common threat. Our findings suggest that the documented plants provide a large resource of AD therapeutic potential. In demonstrating bioactivities targeted to these causal bases, such plants may have the capacity to reduce or reverse AD, with promise as drug leads to target multiple AD hallmarks. However, there is a need to preserve ethnomedical knowledge, and the habitats on which this knowledge depends.
Collapse
Affiliation(s)
| | - Luke D K Tyler
- School of Natural Sciences, Bangor University, Gwynedd, UK
| |
Collapse
|
213
|
Mao M, Wang LY, Zhu LY, Wang F, Ding Y, Tong JH, Sun J, Sun Q, Ji MH. Higher serum PGE2 is a predicative biomarker for postoperative delirium following elective orthopedic surgery in elderly patients. BMC Geriatr 2022; 22:685. [PMID: 35982410 PMCID: PMC9389800 DOI: 10.1186/s12877-022-03367-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background Postoperative delirium (POD), one of the most common complications following major surgery, imposes a heavy burden on patients and society. The objective of this exploratory study was to conduct a secondary analysis to identify whether there exist novel and reliable serum biomarkers for the prediction of POD. Methods A total of 131 adult patients (≥ 65 years) undergoing lower extremity orthopedic surgery with were enrolled in this study. Cognitive function was assessed preoperatively with Mini-Mental State Examination (MMSE). Delirium was diagnosed according to the Confusion Assessment Method (CAM) criteria on preoperative day and postoperative days 1–3. The preoperative serum levels of a panel of 16 biochemical parameters were measured by ELISA. Results Thirty-five patients developed POD, with an incidence of 26.7%. Patients in POD group were older (P = 0.001) and had lower preoperative MMSE scores (P = 0.001). Preoperative serum levels of prostaglandin E2 (PGE2, P < 0.001), S100β (P < 0.001), glial fibrillary acidic protein (P < 0.001) and neurofilament light (P = 0.002) in POD group were significantly increased. Logistic regression analysis showed that advanced age (OR = 1.144, 95%CI: 1.008 ~ 1.298, P = 0.037), higher serum neurofilament light (OR = 1.003, 95%CI: 1.000 ~ 1.005, P = 0.036) and PGE2 (OR = 1.031, 95%CI: 1.018 ~ 1.044, P < 0.001) levels were associated with the development of POD. In addition, serum level of PGE2 yielded an area under the ROC curve (AUC) of 0.897 to predict POD (P < 0.001), with a sensitivity of 80% and a specificity of 83.3%. Conclusions Our study showed that higher preoperative serum PGE2 level might be a biomarker to predict the occurrence of POD in elderly patients undergoing elective orthopedic surgery. Trial registration NCT03792373 www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Meng Mao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Anesthesiology, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lei-Yuan Wang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
| | - Lan-Yue Zhu
- Department of Anesthesiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Fei Wang
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
| | - Ying Ding
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
| | - Jian-Hua Tong
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
| | - Jie Sun
- Department of Anesthesiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Qiang Sun
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Department of Anesthesiology, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Mu-Huo Ji
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China.
| |
Collapse
|
214
|
Early Changes in Transcriptomic Profiles in Synaptodendrosomes Reveal Aberrant Synaptic Functions in Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms23168888. [PMID: 36012153 PMCID: PMC9408306 DOI: 10.3390/ijms23168888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most prevalent neurodegenerative disorders characterized by the progressive decline of cognitive functions, and is closely associated with the dysfunction of synapses, which comprise the basic structure that mediates the communication between neurons. Although the protein architecture and machinery for protein translation at synapses are extensively studied, the impact that local changes in the mRNA reservoir have on AD progression is largely unknown. Here, we investigated the changes in transcriptomic profiles in the synaptodendrosomes purified from the cortices of AD mice at ages 3 and 6 months, a stage when early signatures of synaptic dysfunction are revealed. The transcriptomic profiles of synaptodendrosomes showed a greater number of localized differentially expressed genes (DEGs) in 6-month-old AD mice compared with mice 3 months of age. Gene Ontology (GO) analysis showed that these DEGs are majorly enriched in mitochondrial biogenesis and metabolic activity. More specifically, we further identified three representative DEGs in mitochondrial and metabolic pathways—Prnp, Cst3, and Cox6c—that regulate the dendritic spine density and morphology in neurons. Taken together, this study provides insights into the transcriptomic changes in synaptodendrosomes during AD progression, which may facilitate the development of intervention strategies targeting local translation to ameliorate the pathological progression of AD.
Collapse
|
215
|
Wang Y, Dong C, Han Y, Gu Z, Sun C. Immunosenescence, aging and successful aging. Front Immunol 2022; 13:942796. [PMID: 35983061 PMCID: PMC9379926 DOI: 10.3389/fimmu.2022.942796] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/08/2022] [Indexed: 12/24/2022] Open
Abstract
Aging induces a series of immune related changes, which is called immunosenescence, playing important roles in many age-related diseases, especially neurodegenerative diseases, tumors, cardiovascular diseases, autoimmune diseases and coronavirus disease 2019(COVID-19). However, the mechanism of immunosenescence, the association with aging and successful aging, and the effects on diseases are not revealed obviously. In order to provide theoretical basis for preventing or controlling diseases effectively and achieve successful aging, we conducted the review and found that changes of aging-related phenotypes, deterioration of immune organ function and alterations of immune cell subsets participated in the process of immunosenescence, which had great effects on the occurrence and development of age-related diseases.
Collapse
Affiliation(s)
- Yunan Wang
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chen Dong
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yudian Han
- Information Center, The First People’s Hospital of Nantong City, Nantong, China
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Zhifeng Gu, ; Chi Sun,
| | - Chi Sun
- Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Zhifeng Gu, ; Chi Sun,
| |
Collapse
|
216
|
Gao Y, Wang JB. Commentary: Indirect action pattern: A remote and cross-organ pharmacological mechanism for drug innovation. Acta Pharm Sin B 2022; 12:3448-3450. [PMID: 35967286 PMCID: PMC9366289 DOI: 10.1016/j.apsb.2022.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/19/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Jia-bo Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
217
|
Abstract
Ageing leads to profound alterations in the immune system and increases susceptibility to some chronic, infectious and autoimmune diseases. In recent years, widespread application of single-cell techniques has enabled substantial progress in our understanding of the ageing immune system. These comprehensive approaches have expanded and detailed the current views of ageing and immunity. Here we review a body of recent studies that explored how the immune system ages using unbiased profiling techniques at single-cell resolution. Specifically, we discuss an emergent understanding of age-related alterations in innate and adaptive immune cell populations, antigen receptor repertoires and immune cell-supporting microenvironments of the peripheral tissues. Focusing on the results obtained in mice and humans, we describe the multidimensional data that align with established concepts of immune ageing as well as novel insights emerging from these studies. We further discuss outstanding questions in the field and highlight techniques that will advance our understanding of immune ageing in the future.
Collapse
Affiliation(s)
- Denis A Mogilenko
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Irina Shchukina
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
218
|
Klæstrup IH, Just MK, Holm KL, Alstrup AKO, Romero-Ramos M, Borghammer P, Van Den Berge N. Impact of aging on animal models of Parkinson's disease. Front Aging Neurosci 2022; 14:909273. [PMID: 35966779 PMCID: PMC9366194 DOI: 10.3389/fnagi.2022.909273] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Aging is the biggest risk factor for developing Parkinson's disease (PD), the second most common neurodegenerative disorder. Several animal models have been developed to explore the pathophysiology underlying neurodegeneration and the initiation and spread of alpha-synuclein-related PD pathology, and to investigate biomarkers and therapeutic strategies. However, bench-to-bedside translation of preclinical findings remains suboptimal and successful disease-modifying treatments remain to be discovered. Despite aging being the main risk factor for developing idiopathic PD, most studies employ young animals in their experimental set-up, hereby ignoring age-related cellular and molecular mechanisms at play. Consequently, studies in young animals may not be an accurate reflection of human PD, limiting translational outcomes. Recently, it has been shown that aged animals in PD research demonstrate a higher susceptibility to developing pathology and neurodegeneration, and present with a more disseminated and accelerated disease course, compared to young animals. Here we review recent advances in the investigation of the role of aging in preclinical PD research, including challenges related to aged animal models that are limiting widespread use. Overall, current findings indicate that the use of aged animals may be required to account for age-related interactions in PD pathophysiology. Thus, although the use of older animals has disadvantages, a model that better represents clinical disease within the elderly would be more beneficial in the long run, as it will increase translational value and minimize the risk of therapies failing during clinical studies. Furthermore, we provide recommendations to manage the challenges related to aged animal models.
Collapse
Affiliation(s)
- Ida Hyllen Klæstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- DANDRITE-Danish Research Institute of Translational Neuroscience, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Mie Kristine Just
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | | | - Aage Kristian Olsen Alstrup
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- DANDRITE-Danish Research Institute of Translational Neuroscience, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Per Borghammer
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Nathalie Van Den Berge
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
219
|
Lu X, Yang YM, Lu YQ. Immunosenescence: A Critical Factor Associated With Organ Injury After Sepsis. Front Immunol 2022; 13:917293. [PMID: 35924237 PMCID: PMC9339684 DOI: 10.3389/fimmu.2022.917293] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Progressive immune dysfunction associated with aging is known as immunosenescence. The age-related deterioration of immune function is accompanied by chronic inflammation and microenvironment changes. Immunosenescence can affect both innate and acquired immunity. Sepsis is a systemic inflammatory response that affects parenchymal organs, such as the respiratory system, cardiovascular system, liver, urinary system, and central nervous system, according to the sequential organ failure assessment (SOFA). The initial immune response is characterized by an excess release of inflammatory factors, followed by persistent immune paralysis. Moreover, immunosenescence was found to complement the severity of the immune disorder following sepsis. Furthermore, the immune characteristics associated with sepsis include lymphocytopenia, thymus degeneration, and immunosuppressive cell proliferation, which are very similar to the characteristics of immunosenescence. Therefore, an in-depth understanding of immunosenescence after sepsis and its subsequent effects on the organs may contribute to the development of promising therapeutic strategies. This paper focuses on the characteristics of immunosenescence after sepsis and rigorously analyzes the possible underlying mechanism of action. Based on several recent studies, we summarized the relationship between immunosenescence and sepsis-related organs. We believe that the association between immunosenescence and parenchymal organs might be able to explain the delayed consequences associated with sepsis.
Collapse
Affiliation(s)
- Xuan Lu
- Department of Geriatric and Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- The Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou, China
| | - Yun-Mei Yang
- Department of Geriatric and Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- The Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou, China
| | - Yuan-Qiang Lu
- Department of Geriatric and Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- The Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou, China
- *Correspondence: Yuan-Qiang Lu,
| |
Collapse
|
220
|
Dube CT, Ong YHB, Wemyss K, Krishnan S, Tan TJ, Janela B, Grainger JR, Ronshaugen M, Mace KA, Lim CY. Age-Related Alterations in Macrophage Distribution and Function Are Associated With Delayed Cutaneous Wound Healing. Front Immunol 2022; 13:943159. [PMID: 35874681 PMCID: PMC9304927 DOI: 10.3389/fimmu.2022.943159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Ageing-related delays and dysregulated inflammation in wound healing are well-documented in both human and animal models. However, cellular and molecular changes underlying this impairment in healing progression are not fully understood. In this study, we characterised ageing-associated changes to macrophages in wounds of young and aged mice and investigated transcriptomic differences that may impact the progression of wound healing. Full-thickness wounds created on the dorsum of C57BL/6J young and aged mice were excised on Days 3 and 7 post-wounding for analysis by immunohistochemistry, flow cytometry, and RNA sequencing. Our data revealed that macrophages were significantly reduced in aged wounds in comparison to young. Functional transcriptomic analyses showed that macrophages from aged wounds exhibited significantly reduced expression of cell cycle, DNA replication, and repair pathway genes. Furthermore, we uncovered an elevated pro-inflammatory gene expression program in the aged macrophages correlated with poor inflammation resolution and excessive tissue damage observed in aged wounds. Altogether, our work provides insights into how poorly healing aged wounds are phenotypically defined by the presence of macrophages with reduced proliferative capacity and an exacerbated inflammatory response, both of which are pathways that can be targeted to improve healing in the elderly.
Collapse
Affiliation(s)
- Christabel Thembela Dube
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Epithelial Epigenetics and Development Laboratory, ASTAR Skin Research Labs, Singapore, Singapore
| | - Yasmin Hui Binn Ong
- Epithelial Epigenetics and Development Laboratory, ASTAR Skin Research Labs, Singapore, Singapore
| | - Kelly Wemyss
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Siddharth Krishnan
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Tiak Ju Tan
- Epithelial Epigenetics and Development Laboratory, ASTAR Skin Research Labs, Singapore, Singapore
| | - Baptiste Janela
- Skin Immunology Laboratory, ASTAR Skin Research Labs, Singapore, Singapore
- Skin Immuno-Monitoring Platform , Skin Research Institute of Singapore, Singapore, Singapore
| | - John R. Grainger
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Matthew Ronshaugen
- School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kimberly A. Mace
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Chin Yan Lim
- Epithelial Epigenetics and Development Laboratory, ASTAR Skin Research Labs, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
221
|
Davis CM, Zhang WH, Bah TM, Roese NE, Allen EM, Leung P, Boutros SJ, Marzulla T, Patel E, Nie X, Alkayed FN, Huang JH, Jensen MA, Raber J, Pike MM, Alkayed NJ. Age-dependent cognitive impairment, hydrocephalus and leukocyte infiltration in transgenic mice with endothelial expression of human EPHX2. NPJ AGING 2022; 8:9. [PMID: 35927273 PMCID: PMC9256583 DOI: 10.1038/s41514-022-00090-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 05/31/2022] [Indexed: 12/30/2022]
Abstract
Soluble epoxide hydrolase (sEH) is upregulated in microvascular endothelium of human brain with vascular cognitive impairment (VCI). Transgenic endothelial expression of human sEH in mice (Tie2hsEH) induces endothelial dysfunction (ED), a pathogenetic mechanism of VCI. We sought to determine if endothelial upregulation of sEH is sufficient to cause cognitive impairment, and if cognitive impairment due to chronic hypoperfusion induced by unilateral common carotid artery occlusion (CCAO) is exacerbated in Tie2hsEH mice. Behavioral performance was assessed by the open field, rotarod, novel object, Morris water maze and fear conditioning tests. Cerebral blood flow and brain morphology were evaluated by MRI, and inflammatory changes investigated using immunohistochemistry and flow cytometry. We demonstrate that transgenic endothelial expression of sEH is sufficient to induce cognitive impairment, associated with leukocyte infiltration, brain atrophy and accelerated, age-dependent ventriculomegaly, identifying ED and sEH upregulation as potential underlying mechanisms and therapeutic targets for VCI.
Collapse
Affiliation(s)
- Catherine M Davis
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Wenri H Zhang
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Thierno M Bah
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Natalie E Roese
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Elyse M Allen
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Philberta Leung
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sydney J Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Tessa Marzulla
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Esha Patel
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Xiao Nie
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Farah N Alkayed
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Justin H Huang
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Michael A Jensen
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Martin M Pike
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Nabil J Alkayed
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
222
|
Li D, Huang LT, Zhang CP, Li Q, Wang JH. Insights Into the Role of Platelet-Derived Growth Factors: Implications for Parkinson’s Disease Pathogenesis and Treatment. Front Aging Neurosci 2022; 14:890509. [PMID: 35847662 PMCID: PMC9283766 DOI: 10.3389/fnagi.2022.890509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD), the second most common neurodegenerative disease after Alzheimer’s disease, commonly occurs in the elderly population, causing a significant medical and economic burden to the aging society worldwide. At present, there are few effective methods that achieve satisfactory clinical results in the treatment of PD. Platelet-derived growth factors (PDGFs) and platelet-derived growth factor receptors (PDGFRs) are important neurotrophic factors that are expressed in various cell types. Their unique structures allow for specific binding that can effectively regulate vital functions in the nervous system. In this review, we summarized the possible mechanisms by which PDGFs/PDGFRs regulate the occurrence and development of PD by affecting oxidative stress, mitochondrial function, protein folding and aggregation, Ca2+ homeostasis, and cell neuroinflammation. These modes of action mainly depend on the type and distribution of PDGFs in different nerve cells. We also summarized the possible clinical applications and prospects for PDGF in the treatment of PD, especially in genetic treatment. Recent advances have shown that PDGFs have contradictory roles within the central nervous system (CNS). Although they exert neuroprotective effects through multiple pathways, they are also associated with the disruption of the blood–brain barrier (BBB). Our recommendations based on our findings include further investigation of the contradictory neurotrophic and neurotoxic effects of the PDGFs acting on the CNS.
Collapse
Affiliation(s)
- Dan Li
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng-pu Zhang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiang Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Qiang Li,
| | - Jia-He Wang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- Jia-He Wang,
| |
Collapse
|
223
|
Yahsi B, Gunaydin G. Immunometabolism - The Role of Branched-Chain Amino Acids. Front Immunol 2022; 13:886822. [PMID: 35812393 PMCID: PMC9259854 DOI: 10.3389/fimmu.2022.886822] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Immunometabolism has been the focus of extensive research over the last years, especially in terms of augmenting anti-tumor immune responses. Regulatory T cells (Tregs) are a subset of CD4+ T cells, which have been known for their immunosuppressive roles in various conditions including anti-tumor immune responses. Even though several studies aimed to target Tregs in the tumor microenvironment (TME), such approaches generally result in the inhibition of the Tregs non-specifically, which may cause immunopathologies such as autoimmunity. Therefore, specifically targeting the Tregs in the TME would be vital in terms of achieving a successful and specific treatment. Recently, an association between Tregs and isoleucine, which represents one type of branched-chain amino acids (BCAAs), has been demonstrated. The presence of isoleucine seems to affect majorly Tregs, rather than conventional T cells. Considering the fact that Tregs bear several distinct metabolic features in the TME, targeting their immunometabolic pathways may be a rational approach. In this Review, we provide a general overview on the potential distinct metabolic features of T cells, especially focusing on BCAAs in Tregs as well as in their subtypes.
Collapse
Affiliation(s)
- Berkay Yahsi
- School of Medicine, Hacettepe University, Ankara, Turkey
| | - Gurcan Gunaydin
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
| |
Collapse
|
224
|
Functional MRI Changes in Patients after Thyroidectomy under General Anesthesia. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1935125. [PMID: 35774279 PMCID: PMC9239812 DOI: 10.1155/2022/1935125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/18/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022]
Abstract
Cognitive changes affecting elderly patients following surgery under anesthesia have drawn significant attention and have been investigated in considerable depth. Resting-state functional magnetic resonance imaging (rs-fMRI) can be used to assess changes in brain functional connectivity (FC) associated with postoperative changes in cognition, a common complication in seniors undergoing surgery. In this study, we recruited 20 patients over 55 of age and scheduled an elective thyroidectomy under general anesthesia to assess perioperative changes in brain FC density (FCD) in patients undergoing thyroidectomy under general anesthesia using rs-fMRI. All 20 patients underwent a series of clinical, quantitative, neurological, and neuropsychological tests and fMRI examinations on the day before surgery (Day 0) and 7 days after surgery (Day 7). The following tests were conducted on all patients: the Minimental State Examination (MMSE), the digit symbol substitution test (DSST), the trail making test (part A), the verbal fluency test, and Warrington's recognition memory test (WRMT). FMRI data were acquired using a 3T MR system; the FCD values were calculated using the REST software package. We used paired t-tests to compare the FCD between Day 7 and Day 0. A value of p < 0.05 was considered to reflect statistical significance. The postoperative FCD was significantly reduced in the supplementary motor area (SMA). Analyses of the percentage changes of errors in the WRMT revealed a significant and negative correlation with the mean percentage change of FCD in the SMA (Spearman's r = −0.54, 95% CI: (-0.80, -0.12), p = 0.014). Postoperative changes in FCD in the SMA may be associated with the perioperative neurocognitive changes in patients undergoing partial thyroidectomy under general anesthesia.
Collapse
|
225
|
Liu S, Chen X. MedDiet adherence score for the association between inflammatory markers and cognitive performance in the elderly: a study of the NHANES 2011-2014. BMC Geriatr 2022; 22:511. [PMID: 35729501 PMCID: PMC9215079 DOI: 10.1186/s12877-022-03140-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/03/2022] [Indexed: 11/26/2022] Open
Abstract
Background To explore the optimal Mediterranean diet (MedDiet) adherence score threshold for the association between inflammatory markers and cognitive performance among older adults. Methods In this cross-sectional study, we selected data of the elderly (≥ 60 years old) from the National Health and Nutrition Examination Survey (NHANES) 2011–2014 (n = 2830). Participants completed at least one cognitive measurement and two 24-h food recalls. By analyzing the relation between inflammatory markers and cognitive performance using the subdivided MedDiet adherence score, we got the optimal MedDiet adherence score threshold. Then the optimal threshold was used to divide participants into high and low MedDiet adherence groups and multivariate logistic regression models were developed to examine the association between inflammatory markers and cognitive performance in each group. Subgroup analysis was conducted based on gender, race, BMI, physical activity level, and chronic diseases. Results We chose 4 as the optimal MedDiet adherence score threshold and included these participants whose MedDiet adherence score was 4 or above into the high MedDiet adherence group, while the MedDiet adherence score of the low adherence group was less than 4. We found that the increased white blood cell (WBC) count (OR = 1.44, 95% CI: 1.09–1.90, P = 0.008), neutrophil count (OR = 1.30, 95% CI: 1.03–1.65, P = 0.023), and neutrophil-albumin ratio (NAR) (OR = 1.34, 95% CI: 1.06–1.70, P = 0.012) were all related to a higher risk of low cognitive performance in the low MedDiet adherence group. A higher PLR was linked to a reduced risk of low cognitive performance (OR = 0.86, 95% CI: 0.74–1.00, P = 0.036) in the high MedDiet adherence group. Significant differences were found in the associations of WBC count, neutrophil count and NAR with low cognitive performance between the low and high MedDiet adherence groups (all P < 0.001). The weakened negative association between inflammatory markers and cognitive performance in the high MedDiet adherence group also existed among male, non-Hispanic white, normal-weight, overweight, moderate work activity, moderate recreational activity, non-depression, hypertension, non-hypertension, non-diabetes, non-stroke, non-heart failure, non-coronary heart disease, or non-heart attack subpopulations of older adults. Conclusions The optimal threshold for the MedDiet adherence score was 4, and the negative association between inflammation and cognitive performance could be weakened in older adults whose MedDiet adherence score was ≥ 4. Supplementary Information The online version contains supplementary material available at 10.1186/s12877-022-03140-1.
Collapse
Affiliation(s)
- Shuting Liu
- Internal Medicine Department of the Fifth Outpatient Department, Jinling Hospital Affiliated to Medical School of Nanjing University, Nanjing, 210002, Jiangsu, PR China
| | - Xiaorong Chen
- Neurologic Center, Suining Central Hospital, No.127 Desheng West Road, Chuanshan District, Suining, 629000, Sichuan, PR China.
| |
Collapse
|
226
|
Maheshwari D, Kumar D, Jagdish RK, Nautiyal N, Hidam A, Kumari R, Sehgal R, Trehanpati N, Baweja S, Kumar G, Sinha S, Bajpai M, Pamecha V, Bihari C, Maiwall R, Sarin SK, Kumar A. Bioenergetic Failure Drives Functional Exhaustion of Monocytes in Acute-on-Chronic Liver Failure. Front Immunol 2022; 13:856587. [PMID: 35747140 PMCID: PMC9210982 DOI: 10.3389/fimmu.2022.856587] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
Objective The monocyte–macrophage system is central to the host’s innate immune defense and in resolving injury. It is reported to be dysfunctional in acute-on-chronic liver failure (ACLF). The disease-associated alterations in ACLF monocytes are not fully understood. We investigated the mechanism of monocytes’ functional exhaustion and the role of umbilical cord mesenchymal stem cells (ucMSCs) in re-energizing monocytes in ACLF. Design Monocytes were isolated from the peripheral blood of ACLF patients (n = 34) and matched healthy controls (n = 7) and patients with compensated cirrhosis (n = 7); phagocytic function, oxidative burst, and bioenergetics were analyzed. In the ACLF mouse model, ucMSCs were infused intravenously, and animals were sacrificed at 24 h and day 11 to assess changes in monocyte function, liver injury, and regeneration. Results Patients with ACLF (alcohol 64%) compared with healthy controls and those with compensated cirrhosis had an increased number of peripheral blood monocytes (p < 0.0001) which displayed significant defects in phagocytic (p < 0.0001) and oxidative burst capacity (p < 0.0001). ACLF patients also showed a significant increase in the number of liver macrophages as compared with healthy controls (p < 0.001). Bioenergetic analysis showed markedly reduced oxidative phosphorylation (p < 0.0001) and glycolysis (p < 0.001) in ACLF monocytes. Patients with monocytes having maximum mitochondrial respiration of <37.9 pmol/min [AUC = 0.822, hazard ratio (HR) = 4.5] and baseline glycolysis of ≤42.7 mpH/min (AUC = 0.901, HR = 9.1) showed increased 28-day mortality (p < 0.001). Co-culturing ACLF monocytes with ucMSC showed improved mitochondrial respiration (p < 0.01) and phagocytosis (p < 0.0001). Furthermore, ucMSC therapy increased monocyte energy (p < 0.01) and phagocytosis (p < 0.001), reduced hepatic injury, and enhanced hepatocyte regeneration in ACLF animals. Conclusion Bioenergetic failure drives the functional exhaustion of monocytes in ACLF. ucMSCs resuscitate monocyte energy and prevent its exhaustion. Restoring monocyte function can ameliorate hepatic injury and promote liver regeneration in the animal model of ACLF.
Collapse
Affiliation(s)
- Deepanshu Maheshwari
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Dhananjay Kumar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rakesh Kumar Jagdish
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Nidhi Nautiyal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Ashinikumar Hidam
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rekha Kumari
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rashi Sehgal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Nirupama Trehanpati
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Sukriti Baweja
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Guresh Kumar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Swati Sinha
- Department of Obstetrics and Gynaecology, Sitaram Bhartia Institute of Science and Research, New Delhi, India
| | - Meenu Bajpai
- Department of Transfusion Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Viniyendra Pamecha
- Department of Hepato-Pancreato-Biliary (HPB) Surgery and Liver Transplant, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Chhagan Bihari
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rakhi Maiwall
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
- *Correspondence: Anupam Kumar, ; Shiv Kumar Sarin,
| | - Anupam Kumar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
- *Correspondence: Anupam Kumar, ; Shiv Kumar Sarin,
| |
Collapse
|
227
|
Fields ME, Mirro AE, Binkley MM, Guilliams KP, Lewis JB, Fellah S, Chen Y, Hulbert ML, An H, Ford AL, Lee J. Cerebral oxygen metabolic stress is increased in children with sickle cell anemia compared to anemic controls. Am J Hematol 2022; 97:682-690. [PMID: 35113471 DOI: 10.1002/ajh.26485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/22/2022] [Accepted: 01/29/2022] [Indexed: 01/28/2023]
Abstract
Patients with sickle cell anemia (SCA) experience cerebral metabolic stress with an increase in oxygen extraction fraction (OEF) to compensate for reduced oxygen carrying capacity due to anemia. It remains unclear if anemia alone drives this metabolic stress. Using MRI, we collected voxel-wise OEF measurements to test our hypothesis that OEF would be elevated in anemic controls without SCA (AC) compared to healthy controls (HC), but OEF would be even higher in SCA compared to AC. Brain MRIs (N = 159) were obtained in 120 participants (34 HC, 27 AC, 59 SCA). While hemoglobin was lower in AC versus HC (p < 0.001), hemoglobin was not different between AC and SCA cohorts (p = 0.459). Whole brain OEF was higher in AC compared to HC (p < 0.001), but lower compared to SCA (p = 0.001). Whole brain OEF remained significantly higher in SCA compared to HC (p = 0.001) while there was no longer a difference between AC versus HC (p = 0.935) in a multivariate model controlling for age and hemoglobin. OEF peaked within the border zone regions of the brain in both SCA and AC cohorts, but the volume of white matter with regionally elevated OEF in AC was smaller (1.8%) than SCA (58.0%). While infarcts colocalized within regions of elevated OEF, more SCA participants had infarcts than AC (p < 0.001). We conclude that children with SCA experience elevated OEF compared to AC and HC after controlling for the impact of anemia, suggesting that there are other pathophysiologic factors besides anemia contributing to cerebral metabolic stress in children with SCA.
Collapse
Affiliation(s)
- Melanie E. Fields
- Department of Pediatrics Washington University School of Medicine St. Louis Missouri USA
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
| | - Amy E. Mirro
- Department of Pediatrics Washington University School of Medicine St. Louis Missouri USA
| | - Michael M. Binkley
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
| | - Kristin P. Guilliams
- Department of Pediatrics Washington University School of Medicine St. Louis Missouri USA
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
- Mallinckrodt Institute of Radiology Washington University School of Medicine St. Louis Missouri USA
| | - Josiah B. Lewis
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
| | - Slim Fellah
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
| | - Yasheng Chen
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
| | - Monica L. Hulbert
- Department of Pediatrics Washington University School of Medicine St. Louis Missouri USA
| | - Hongyu An
- Mallinckrodt Institute of Radiology Washington University School of Medicine St. Louis Missouri USA
| | - Andria L. Ford
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
- Mallinckrodt Institute of Radiology Washington University School of Medicine St. Louis Missouri USA
| | - Jin‐Moo Lee
- Department of Neurology Washington University School of Medicine St. Louis Missouri USA
- Mallinckrodt Institute of Radiology Washington University School of Medicine St. Louis Missouri USA
- Department of Biomedical Engineering Washington University School of Medicine St. Louis Missouri USA
| |
Collapse
|
228
|
Schwartz M, Cahalon L. The vicious cycle governing the brain–immune system relationship in neurodegenerative diseases. Curr Opin Immunol 2022; 76:102182. [DOI: 10.1016/j.coi.2022.102182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022]
|
229
|
Yang C, Pang Y, Huang Y, Ye F, Chen X, Gao Y, Zhang C, Yao L, Gao J. Single-cell transcriptomics identifies premature aging features of TERC-deficient mouse brain and bone marrow. GeroScience 2022; 44:2139-2155. [PMID: 35545739 DOI: 10.1007/s11357-022-00578-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/22/2022] [Indexed: 11/29/2022] Open
Abstract
Aging is a progressive loss of physiological function and increased susceptibility to major pathologies. Degenerative diseases in both brain and bone including Alzheimer disease (AD) and osteoporosis are common in aging groups. TERC is RNA component of telomerase, and its deficiency accelerates aging-related phenotypes including impaired life span, organ failure, bone loss, and brain dysfunction. In this study, we investigated the traits of bone marrow-brain cross-tissue communications in young mice, natural aging mice, and premature aging (TERC deficient, TERC-KO) mice by single-cell transcriptome sequencing. Differentially expressed gene analysis of brain as well as bone marrow between premature aging mouse and young mouse demonstrated aging-related inflammatory response and suppression of neuron development. Further analysis of senescence-associated secretory phenotype (SASP) landscape indicated that TERC-KO perturbation was enriched in oligodendrocyte progenitor cells (OPCs) and hematopoietic stem and progenitor cells (HSPC). Series of inflammatory associated myeloid cells was activated in premature aging mice brain and bone marrow. Cross-tissue comparison of TERC-KO mice brain and bone marrow illustrated obvious ligand-receptor communications between brain glia cells, macrophages, and bone marrow myeloid cells in premature aging-induced inflammation. Enrichment of co-regulation modules between brain and bone marrow identified premature aging response genes such as Dusp1 and Ifitm3. Our study provides a rich resource for understanding premature aging-associated perturbation in brain and bone marrow and supporting myeloid cells and endothelial cells as promising therapy targeting for age-related brain-bone diseases.
Collapse
Affiliation(s)
- Chunying Yang
- Department of Neurology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yigang Huang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Fang Ye
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiaoyi Chen
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315000, Zhejiang, China
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Lufeng Yao
- Department of Orthopaedic Surgery, Ningbo No. 6 Hospital, Ningbo, 315040, Zhejiang, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
230
|
Mone P, Pansini A, Jankauskas SS, Varzideh F, Kansakar U, Lombardi A, Trimarco V, Frullone S, Santulli G. L-Arginine Improves Cognitive Impairment in Hypertensive Frail Older Adults. Front Cardiovasc Med 2022; 9:868521. [PMID: 35498050 PMCID: PMC9039514 DOI: 10.3389/fcvm.2022.868521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
Cognitive impairment is a prevailing event in hypertensive patients and in frail older adults. Endothelial dysfunction has been shown to underlie both hypertension and cognitive dysfunction. Our hypothesis is that L-Arginine, which is known to ameliorate endothelial dysfunction, could counteract cognitive impairment in a high-risk population of hypertensive frail older adults. We designed a clinical trial to verify the effects of 4-weeks oral supplementation of L-Arginine on global cognitive function of hypertensive frail older patients. The study was successfully completed by 35 frail hypertensive elderly patients assigned to L-Arginine and 37 assigned to placebo. At follow-up, we found a significant difference in the Montreal Cognitive Assessment (MoCA) test score between the L-Arginine treated group and placebo (p: 0.0178). Moreover, we demonstrated that L-Arginine significantly attenuates Angiotensin II-induced mitochondrial oxidative stress in human endothelial cells. In conclusion, our findings indicate for the first time that oral L-Arginine supplementation significantly improves cognitive impairment in frail hypertensive older adults.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States.,Azienda Sanitaria Locale (ASL) Avellino, Avellino, Italy.,Campania University, Naples, Italy
| | | | | | - Fahimeh Varzideh
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Urna Kansakar
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Angela Lombardi
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | | | | | - Gaetano Santulli
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States.,University of Naples "Federico II", Naples, Italy
| |
Collapse
|
231
|
Willis CM, Nicaise AM, Krzak G, Ionescu RB, Pappa V, D'Angelo A, Agarwal R, Repollés-de-Dalmau M, Peruzzotti-Jametti L, Pluchino S. Soluble factors influencing the neural stem cell niche in brain physiology, inflammation, and aging. Exp Neurol 2022; 355:114124. [DOI: 10.1016/j.expneurol.2022.114124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/16/2022] [Accepted: 05/21/2022] [Indexed: 11/27/2022]
|
232
|
Burtscher J, Romani M, Bernardo G, Popa T, Ziviani E, Hummel FC, Sorrentino V, Millet GP. Boosting mitochondrial health to counteract neurodegeneration. Prog Neurobiol 2022; 215:102289. [DOI: 10.1016/j.pneurobio.2022.102289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 12/22/2022]
|
233
|
Cai Y, Liu J, Wang B, Sun M, Yang H. Microglia in the Neuroinflammatory Pathogenesis of Alzheimer's Disease and Related Therapeutic Targets. Front Immunol 2022; 13:856376. [PMID: 35558075 PMCID: PMC9086828 DOI: 10.3389/fimmu.2022.856376] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide, characterized by progressive neuron degeneration or loss due to excessive accumulation of β-amyloid (Aβ) peptides, formation of neurofibrillary tangles (NFTs), and hyperphosphorylated tau. The treatment of AD has been only partially successful as the majority of the pharmacotherapies on the market may alleviate some of the symptoms. In the occurrence of AD, increasing attention has been paid to neurodegeneration, while the resident glial cells, like microglia are also observed. Microglia, a kind of crucial glial cells associated with the innate immune response, functions as double-edge sword role in CNS. They exert a beneficial or detrimental influence on the adjacent neurons through secretion of both pro-inflammatory cytokines as well as neurotrophic factors. In addition, their endocytosis of debris and toxic protein like Aβ and tau ensures homeostasis of the neuronal microenvironment. In this review, we will systematically summarize recent research regarding the roles of microglia in AD pathology and latest microglia-associated therapeutic targets mainly including pro-inflammatory genes, anti-inflammatory genes and phagocytosis at length, some of which are contradictory and controversial and warrant to further be investigated.
Collapse
Affiliation(s)
| | | | | | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
234
|
Stearoyl-CoA Desaturase inhibition reverses immune, synaptic and cognitive impairments in an Alzheimer's disease mouse model. Nat Commun 2022; 13:2061. [PMID: 35443751 PMCID: PMC9021296 DOI: 10.1038/s41467-022-29506-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
The defining features of Alzheimer’s disease (AD) include alterations in protein aggregation, immunity, lipid metabolism, synapses, and learning and memory. Of these, lipid abnormalities are the least understood. Here, we investigate the role of Stearoyl-CoA desaturase (SCD), a crucial regulator of fatty acid desaturation, in AD pathogenesis. We show that inhibiting brain SCD activity for 1-month in the 3xTg mouse model of AD alters core AD-related transcriptomic pathways in the hippocampus, and that it concomitantly restores essential components of hippocampal function, including dendritic spines and structure, immediate-early gene expression, and learning and memory itself. Moreover, SCD inhibition dampens activation of microglia, key mediators of spine loss during AD and the main immune cells of the brain. These data reveal that brain fatty acid metabolism links AD genes to downstream immune, synaptic, and functional impairments, identifying SCD as a potential target for AD treatment. Alzheimer’s disease (AD) is characterized by lipid abnormalities which are not well understood. Here, the authors investigate the role of Stearoyl-CoA desaturase (SCD) in a mouse model of AD. They show that inhibiting SCD activity induces major brain and immune cell transcriptional changes and restores dendritic structure and learning and memory.
Collapse
|
235
|
Wang J, Xia Z, Sheng P, Rui Y, Cao J, Zhang J, Gao M, Wang L, Yu D, Yan BC. Targeting MicroRNA-144/451-AKT-GSK3β Axis Affects the Proliferation and Differentiation of Radial Glial Cells in the Mouse Hippocampal Dentate Gyrus. ACS Chem Neurosci 2022; 13:897-909. [PMID: 35261236 DOI: 10.1021/acschemneuro.1c00636] [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: 11/30/2022] Open
Abstract
It is well known that aging induces a progressive decline in the proliferation and neural differentiation of radial glial cells (RGCs) in the hippocampal dentate gyrus (DG). The function of miR-144/451 is to activate stress-regulated molecular gene expression switches for cell proliferation and differentiation. We found that the miR-144/451 expression in the hippocampus was significantly reduced in aged mice compared to adult mice. Furthermore, the proliferation and neural differentiation of RGCs in the mouse hippocampal DG was decreased by miR-144/451 knockout (miR-144/451-/-). Antioxidant agents, superoxide dismutases (SODs) and catalase, and the expression of melatonin's receptor in the hippocampus were decreased in the miR-144/451-/- mice. In addition, the (protein kinase B) AKT/(glycogen synthase kinase 3β) GSK3β/(catenin beta-1) β-catenin signaling pathway was weakly activated in the hippocampus of miR-144/451-/- mice, which was related to brain neurogenesis. Melatonin treatment improved the expression of miR-144/451 and antioxidant enzymes and activated the AKT/GSK3β/β-catenin pathway in the hippocampus of miR-144/451-/- mice. When the AKT pathway was inhibited by LY294002, the neurogenerative and antioxidant effects of melatonin were significantly limited in the hippocampus of miR-144/451-/- mice. In brief, our results indicated that miR-144/451 plays crucial roles in the proliferation and neural differentiation of RGCs via the regulation of the antioxidant and AKT/GSK3β/β-catenin pathways.
Collapse
Affiliation(s)
- Jie Wang
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zihao Xia
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Peng Sheng
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Yanggang Rui
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Jianwen Cao
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Jie Zhang
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Manman Gao
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Li Wang
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| | - Duonan Yu
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou 225009, China
| | - Bing Chun Yan
- Medical College, Institute of Translational Medicine, Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University, Yangzhou 225001, PR China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| |
Collapse
|
236
|
Han G, Zhen W, Dai Y, Yu H, Li D, Ma T. Dihuang-Yinzi Alleviates Cognition Deficits via Targeting Energy-Related Metabolism in an Alzheimer Mouse Model as Demonstrated by Integration of Metabolomics and Network Pharmacology. Front Aging Neurosci 2022; 14:873929. [PMID: 35431901 PMCID: PMC9011333 DOI: 10.3389/fnagi.2022.873929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
Energy metabolism disturbance and the consequent reactive oxygen species (ROS) overproduction play a key and pathogenic role in the onset and progression of Alzheimer’s disease (AD). Dihuang-Yinzi (DHYZ) is a traditional Chinese herbal prescription clinically applied to treat AD and other neurodegenerative diseases for a long time. However, the systematical metabolic mechanism of DHYZ against AD remains largely unclear. Here we aimed to explore the mechanism of DHYZ in the treatment of AD comprehensively in an in vivo metabolic context by performing metabolomics analysis coupled with network pharmacology study and experimental validation. The network pharmacology was applied to dig out the potential target of DHYZ against AD. The metabolomics analysis based on UPLC-HRMS was carried out to profile the urine of 2× Tg-AD mice treated with DHYZ. By integrating network pharmacology and metabolomics, we found DHYZ could ameliorate 4 key energy-related metabolic pathways, including glycerophospholipid metabolism, nicotinate/nicotinamide metabolism, glycolysis, and tricarboxylic acid cycle. Besides, we identified 5 potential anti-AD targets of DHYZ, including DAO, HIF1A, PARP1, ALDH3B2, and ACHE, and 14 key differential metabolites involved in the 4 key energy-related metabolic pathways. Furthermore, DHYZ depressed the mitochondrial dysfunction and the resultant ROS overproduction through ameliorating glycerophospholipid metabolism disturbance. Thereby DHYZ increased nicotinamide adenine dinucleotide (NAD+) content and promoted glycolysis and tricarboxylic acid (TCA) cycle, and consequently improved oxidative phosphorylation and energy metabolism. In the present study, we provided a novel, comprehensive and systematic insight into investigating the therapeutic efficacy of DHYZ against AD via ameliorating energy-related metabolism.
Collapse
Affiliation(s)
- Guanghui Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weizhe Zhen
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Dai
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongni Yu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongyue Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Ma
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Tao Ma,
| |
Collapse
|
237
|
Yuan Y, Fan G, Liu Y, Liu L, Zhang T, Liu P, Tu Q, Zhang X, Luo S, Yao L, Chen F, Li J. The transcription factor KLF14 regulates macrophage glycolysis and immune function by inhibiting HK2 in sepsis. Cell Mol Immunol 2022; 19:504-515. [PMID: 34983946 PMCID: PMC8976055 DOI: 10.1038/s41423-021-00806-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/10/2022] Open
Abstract
Sepsis is a heterogeneous syndrome induced by a dysregulated host response to infection. Glycolysis plays a role in maintaining the immune function of macrophages, which is crucial for severely septic patients. However, how the pathways that link glycolysis and macrophages are regulated is still largely unknown. Here, we provide evidence to support the function of KLF14, a novel Krüppel-like transcription factor, in the regulation of glycolysis and the immune function of macrophages during sepsis. KLF14 deletion led to significantly increased mortality in lethal models of murine endotoxemia and sepsis. Mechanistically, KLF14 decreased glycolysis and the secretion of inflammatory cytokines by macrophages by inhibiting the transcription of HK2. In addition, we confirmed that the expression of KLF14 was upregulated in septic patients. Furthermore, pharmacological activation of KLF14 conferred protection against sepsis in mice. These findings uncover a key role of KLF14 in modulating the inflammatory signaling pathway and shed light on the development of KLF14-targeted therapeutics for sepsis.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Guangjian Fan
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Lu Liu
- Department of Anesthesiology, Weifang Medical University, Weifang, 261000, China
| | - Tong Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Pengfei Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Qing Tu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Xinyi Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China
| | - Shiyuan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Liangfang Yao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Feng Chen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Jingbao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 201620, China.
| |
Collapse
|
238
|
Mora I, Arola L, Caimari A, Escoté X, Puiggròs F. Structured Long-Chain Omega-3 Fatty Acids for Improvement of Cognitive Function during Aging. Int J Mol Sci 2022; 23:3472. [PMID: 35408832 PMCID: PMC8998232 DOI: 10.3390/ijms23073472] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023] Open
Abstract
Although the human lifespan has increased in the past century owing to advances in medicine and lifestyle, the human healthspan has not kept up the same pace, especially in brain aging. Consequently, the role of preventive health interventions has become a crucial strategy, in particular, the identification of nutritional compounds that could alleviate the deleterious effects of aging. Among nutrients to cope with aging in special cognitive decline, the long-chain omega-3 polyunsaturated fatty acids (ω-3 LCPUFAs) docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), have emerged as very promising ones. Due to their neuroinflammatory resolving effects, an increased status of DHA and EPA in the elderly has been linked to better cognitive function and a lower risk of dementia. However, the results from clinical studies do not show consistent evidence and intake recommendations for old adults are lacking. Recently, supplementation with structured forms of EPA and DHA, which can be derived natural forms or targeted structures, have proven enhanced bioavailability and powerful benefits. This review summarizes present and future perspectives of new structures of ω-3 LCPUFAs and the role of "omic" technologies combined with the use of high-throughput in vivo models to shed light on the relationships and underlying mechanisms between ω-3 LCPUFAs and healthy aging.
Collapse
Affiliation(s)
- Ignasi Mora
- Brudy Technology S.L., 08006 Barcelona, Spain
| | - Lluís Arola
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| | - Xavier Escoté
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health Unit, 43204 Reus, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| |
Collapse
|
239
|
Ahn K, Lee SJ, Mook-Jung I. White matter-associated microglia: New players in brain aging and neurodegenerative diseases. Ageing Res Rev 2022; 75:101574. [PMID: 35093614 DOI: 10.1016/j.arr.2022.101574] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022]
Abstract
There has been growing interest in brain aging and rejuvenation. It is well known that brain aging is one of the leading causes of neurodegenerative diseases, such as Alzheimer's disease, but brain aging alone can cause cognitive decline. Microglia are thought to act as 'conductors' of white matter aging by modulating diverse glial cells and phagocytosing white matter-derived myelin debris. A recent study identified a specific subpopulation of microglia in the white matter of aged mice, termed white matter-associated microglia (WAM). Additionally, senescent microglia show impaired phagocytic function and altered lipid metabolism, which cause accumulation of lipid metabolites and eventually lead to myelin sheath degeneration. These results suggest that senescent WAM could be pivotal players in axonal loss during brain aging. The aim of this review is to assess the current state of knowledge on brain aging, with an emphasis on the roles of the white matter and microglia, and suggest potential approaches for rejuvenating the aged brain.
Collapse
Affiliation(s)
- Kyusik Ahn
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
240
|
Wculek SK, Dunphy G, Heras-Murillo I, Mastrangelo A, Sancho D. Metabolism of tissue macrophages in homeostasis and pathology. Cell Mol Immunol 2022; 19:384-408. [PMID: 34876704 PMCID: PMC8891297 DOI: 10.1038/s41423-021-00791-9] [Citation(s) in RCA: 212] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023] Open
Abstract
Cellular metabolism orchestrates the intricate use of tissue fuels for catabolism and anabolism to generate cellular energy and structural components. The emerging field of immunometabolism highlights the importance of cellular metabolism for the maintenance and activities of immune cells. Macrophages are embryo- or adult bone marrow-derived leukocytes that are key for healthy tissue homeostasis but can also contribute to pathologies such as metabolic syndrome, atherosclerosis, fibrosis or cancer. Macrophage metabolism has largely been studied in vitro. However, different organs contain diverse macrophage populations that specialize in distinct and often tissue-specific functions. This context specificity creates diverging metabolic challenges for tissue macrophage populations to fulfill their homeostatic roles in their particular microenvironment and conditions their response in pathological conditions. Here, we outline current knowledge on the metabolic requirements and adaptations of macrophages located in tissues during homeostasis and selected diseases.
Collapse
Affiliation(s)
- Stefanie K Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| | - Gillian Dunphy
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Ignacio Heras-Murillo
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Annalaura Mastrangelo
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
241
|
Zhou WJ, Yang HL, Mei J, Chang KK, Lu H, Lai ZZ, Shi JW, Wang XH, Wu K, Zhang T, Wang J, Sun JS, Ye JF, Li DJ, Zhao JY, Jin LP, Li MQ. Fructose-1,6-bisphosphate prevents pregnancy loss by inducing decidual COX-2 + macrophage differentiation. SCIENCE ADVANCES 2022; 8:eabj2488. [PMID: 35196096 PMCID: PMC8865779 DOI: 10.1126/sciadv.abj2488] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 12/23/2021] [Indexed: 05/23/2023]
Abstract
Decidualization is an intricate biological process in which extensive remodeling of the endometrium occurs to support the development of an implanting blastocyst. However, the immunometabolic mechanisms underlying this process are still largely unknown. We found that the decidualization process is accompanied by the accumulation of fructose-1,6-bisphosphate (FBP). The combination of FBP with pyruvate kinase M stimulated IL-27 secretion by endometrial stromal cells in an ERK/c-FOS-dependent manner. IL-27 induced decidual COX-2+ M2-like macrophage differentiation, which promotes decidualization, trophoblast invasion, and maternal-fetal tolerance. Transfer of Ptgs2+/COX-2+ macrophages prevented fetal loss in Il27ra-deleted pregnant mice. FBP levels were low in plasma and decidual tissues of patients with unexplained recurrent spontaneous abortion. In therapeutic studies, FBP supplementation significantly improved embryo loss by up-regulation of IL-27-induced COX-2+ macrophage differentiation in a mouse model of spontaneous abortion. These findings collectively provide a scientific basis for a potential therapeutic strategy to prevent pregnancy loss.
Collapse
Affiliation(s)
- Wen-Jie Zhou
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People’s Republic of China
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Hui-Li Yang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jie Mei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medicine School, Nanjing 210000, People’s Republic of China
| | - Kai-Kai Chang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Han Lu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Zhen-Zhen Lai
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jia-Wei Shi
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Xiao-Hui Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Ke Wu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, People’s Republic of China
| | - Jian Wang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jian-Song Sun
- National Research Centre for Carbohydrate Synthesis, Jiangxi Normal University, Nanchang, Jiangxi Province 330022, People’s Republic of China
| | - Jiang-Feng Ye
- Division of Obstetrics and Gynecology, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jian-Yuan Zhao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, People’s Republic of China
| | - Li-Ping Jin
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| |
Collapse
|
242
|
Shirato K, Sato S. Macrophage Meets the Circadian Clock: Implication of the Circadian Clock in the Role of Macrophages in Acute Lower Respiratory Tract Infection. Front Cell Infect Microbiol 2022; 12:826738. [PMID: 35281442 PMCID: PMC8904936 DOI: 10.3389/fcimb.2022.826738] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
The circadian rhythm is a biological system that creates daily variations of physiology and behavior with a 24-h cycle, which is precisely controlled by the molecular circadian clock. The circadian clock dominates temporal activity of physiological homeostasis at the molecular level, including endocrine secretion, metabolic, immune response, coupled with extrinsic environmental cues (e.g., light/dark cycles) and behavioral cues (e.g., sleep/wake cycles and feeding/fasting cycles). The other side of the clock is that the misaligned circadian rhythm contributes to the onset of a variety of diseases, such as cancer, metabolic diseases, and cardiovascular diseases, the acceleration of aging, and the development of systemic inflammation. The role played by macrophages is a key mediator between circadian disruption and systemic inflammation. At the molecular level, macrophage functions are under the direct control of the circadian clock, and thus the circadian misalignment remodels the phenotype of macrophages toward a ‘killer’ mode. Remarkably, the inflammatory macrophages induce systemic and chronic inflammation, leading to the development of inflammatory diseases and the dampened immune defensive machinery against infectious diseases such as COVID-19. Here, we discuss how the circadian clock regulates macrophage immune functions and provide the potential risk of misaligned circadian rhythms against inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Ken Shirato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University School of Medicine, Mitaka, Japan
| | - Shogo Sato
- Center for Biological Clocks Research, Department of Biology, Texas A&M University, College Station, TX, United States
- *Correspondence: Shogo Sato,
| |
Collapse
|
243
|
Wallis ZK, Williams KC. Monocytes in HIV and SIV Infection and Aging: Implications for Inflamm-Aging and Accelerated Aging. Viruses 2022; 14:409. [PMID: 35216002 PMCID: PMC8880456 DOI: 10.3390/v14020409] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 12/11/2022] Open
Abstract
Before the antiretroviral therapy (ART) era, people living with HIV (PLWH) experienced complications due to AIDS more so than aging. With ART and the extended lifespan of PLWH, HIV comorbidities also include aging-most likely due to accelerated aging-as well as a cardiovascular, neurocognitive disorders, lung and kidney disease, and malignancies. The broad evidence suggests that HIV with ART is associated with accentuated aging, and that the age-related comorbidities occur earlier, due in part to chronic immune activation, co-infections, and possibly the effects of ART alone. Normally the immune system undergoes alterations of lymphocyte and monocyte populations with aging, that include diminished naïve T- and B-lymphocyte numbers, a reliance on memory lymphocytes, and a skewed production of myeloid cells leading to age-related inflammation, termed "inflamm-aging". Specifically, absolute numbers and relative proportions of monocytes and monocyte subpopulations are skewed with age along with myeloid mitochondrial dysfunction, resulting in increased accumulation of reactive oxygen species (ROS). Additionally, an increase in biomarkers of myeloid activation (IL-6, sCD14, and sCD163) occurs with chronic HIV infection and with age, and may contribute to immunosenescence. Chronic HIV infection accelerates aging; meanwhile, ART treatment may slow age-related acceleration, but is not sufficient to stop aging or age-related comorbidities. Overall, a better understanding of the mechanisms behind accentuated aging with HIV and the effects of myeloid activation and turnover is needed for future therapies.
Collapse
|
244
|
Ljungblad L, Bergqvist F, Tümmler C, Madawala S, Olsen TK, Andonova T, Jakobsson PJ, Johnsen JI, Pickova J, Strandvik B, Kogner P, Gleissman H, Wickström M. Omega-3 fatty acids decrease CRYAB, production of oncogenic prostaglandin E 2 and suppress tumor growth in medulloblastoma. Life Sci 2022; 295:120394. [PMID: 35157910 DOI: 10.1016/j.lfs.2022.120394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 12/09/2022]
Abstract
AIMS Medulloblastoma (MB) is one of the most common malignant central nervous system tumors of childhood. Despite intensive treatments that often leads to severe neurological sequelae, the risk for resistant relapses remains significant. In this study we have evaluated the effects of the ω3-long chain polyunsaturated fatty acids (ω3-LCPUFA) docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on MB cell lines and in a MB xenograft model. MAIN METHODS Effects of ω3-LCPUFA treatment of MB cells were assessed using the following: WST-1 assay, cell death probes, clonogenic assay, ELISA and western blot. MB cells were implanted into nude mice and the mice were randomized to DHA, or a combination of DHA and EPA treatment, or to control group. Treatment effects in tumor tissues were evaluated with: LC-MS/MS, RNA-sequencing and immunohistochemistry, and tumors, erythrocytes and brain tissues were analyzed with gas chromatography. KEY FINDINGS ω3-LCPUFA decreased prostaglandin E2 (PGE2) secretion from MB cells, and impaired MB cell viability and colony forming ability and increased apoptosis in a dose-dependent manner. DHA reduced tumor growth in vivo, and both PGE2 and prostacyclin were significantly decreased in tumor tissue from treated mice compared to control animals. All ω3-LCPUFA and dihomo-γ-linolenic acid increased in tumors from treated mice. RNA-sequencing revealed 10 downregulated genes in common among ω3-LCPUFA treated tumors. CRYAB was the most significantly altered gene and the downregulation was confirmed by immunohistochemistry. SIGNIFICANCE Our findings suggest that addition of DHA and EPA to the standard MB treatment regimen might be a novel approach to target inflammation in the tumor microenvironment.
Collapse
Affiliation(s)
- Linda Ljungblad
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| | - Filip Bergqvist
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Conny Tümmler
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Samanthi Madawala
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Thale Kristin Olsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Teodora Andonova
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Jana Pickova
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Birgitta Strandvik
- Department of Biosciences and Nutrition Karolinska Institutet, NEO, Flemingsberg, Stockholm, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Pediatric Oncology, Astrid Lindgrens Childrens Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Helena Gleissman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
245
|
Aron L, Zullo J, Yankner BA. The adaptive aging brain. Curr Opin Neurobiol 2022; 72:91-100. [PMID: 34689041 PMCID: PMC8901453 DOI: 10.1016/j.conb.2021.09.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/12/2021] [Indexed: 01/22/2023]
Abstract
The aging brain is shaped by many structural and functional alterations. Recent cross-disciplinary efforts have uncovered powerful and integrated adaptive mechanisms that promote brain health and prevent functional decline during aging. Here, we review some of the most robust adaptive mechanisms and how they can be engaged to protect, and restore the aging brain.
Collapse
Affiliation(s)
- Liviu Aron
- Department of Genetics, Harvard Medical School, Boston, 02115, MA, USA
| | - Joseph Zullo
- Department of Genetics, Harvard Medical School, Boston, 02115, MA, USA
| | - Bruce A Yankner
- Department of Genetics, Harvard Medical School, Boston, 02115, MA, USA.
| |
Collapse
|
246
|
Adoptive transfer of metabolically reprogrammed macrophages for atherosclerosis treatment in diabetic ApoE−/- mice. Bioact Mater 2022; 16:82-94. [PMID: 35386323 PMCID: PMC8958426 DOI: 10.1016/j.bioactmat.2022.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 01/02/2023] Open
Abstract
Atherosclerosis is characterized by inflammation in the arterial wall, which is known to be exacerbated by diabetes. Therapeutic repression of inflammation is a promising strategy for treating atherosclerosis. In this study, we showed that diabetes aggravated atherosclerosis in apolipoproteinE knockout (ApoE−/-) mice, in which increased expression of long-chain acyl-CoA synthetase 1 (Acsl1) in macrophages played an important role. Knockdown of Acsl1 in macrophages (MφshAcsl1) reprogrammed macrophages to an anti-inflammatory phenotype, especially under hyperglycemic conditions. Injection of MφshAcsl1 reprogrammed macrophages into streptozotocin (STZ)-induced diabetic ApoE−/- mice (ApoE−/-+ STZ) alleviated inflammation locally in the plaque, liver and spleen. Consistent with the reduction in inflammation, plaques became smaller and more stable after the adoptive transfer of reprogrammed macrophages. Taken together, our findings indicate that increased Acsl1 expression in macrophages play a key role in aggravated atherosclerosis of diabetic mice, possibly by promoting inflammation. Adoptive transfer of Acsl1 silenced macrophages may serve as a potential therapeutic strategy for atherosclerosis. Increased Acsl1 in macrophages is responsible for the exacerbated inflammation in diabetes MφshAcsl1 is characterized as anti-inflammatory phenotype Adoptive transfer of MφshAcsl1 alleviates atherosclerosis in diabetic ApoE−/- mice MφshAcsl1 inhibits both local and systemic inflammation in vivo
Collapse
|
247
|
Johnson ECB, Carter EK, Dammer EB, Duong DM, Gerasimov ES, Liu Y, Liu J, Betarbet R, Ping L, Yin L, Serrano GE, Beach TG, Peng J, De Jager PL, Haroutunian V, Zhang B, Gaiteri C, Bennett DA, Gearing M, Wingo TS, Wingo AP, Lah JJ, Levey AI, Seyfried NT. Large-scale deep multi-layer analysis of Alzheimer's disease brain reveals strong proteomic disease-related changes not observed at the RNA level. Nat Neurosci 2022; 25:213-225. [PMID: 35115731 PMCID: PMC8825285 DOI: 10.1038/s41593-021-00999-y] [Citation(s) in RCA: 289] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 12/08/2021] [Indexed: 12/16/2022]
Abstract
The biological processes that are disrupted in the Alzheimer's disease (AD) brain remain incompletely understood. In this study, we analyzed the proteomes of more than 1,000 brain tissues to reveal new AD-related protein co-expression modules that were highly preserved across cohorts and brain regions. Nearly half of the protein co-expression modules, including modules significantly altered in AD, were not observed in RNA networks from the same cohorts and brain regions, highlighting the proteopathic nature of AD. Two such AD-associated modules unique to the proteomic network included a module related to MAPK signaling and metabolism and a module related to the matrisome. The matrisome module was influenced by the APOE ε4 allele but was not related to the rate of cognitive decline after adjustment for neuropathology. By contrast, the MAPK/metabolism module was strongly associated with the rate of cognitive decline. Disease-associated modules unique to the proteome are sources of promising therapeutic targets and biomarkers for AD.
Collapse
Affiliation(s)
- Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| | - E Kathleen Carter
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M Duong
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Yue Liu
- Department of Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jiaqi Liu
- Department of Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ranjita Betarbet
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lingyan Ping
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Luming Yin
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Taub Institute, Columbia University Irving Medical Center, New York Presbyterian Hospital, New York, NY, USA
| | - Vahram Haroutunian
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center MIRECC, Bronx, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Marla Gearing
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas S Wingo
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Aliza P Wingo
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
- Division of Mental Health, Atlanta VA Medical Center, Atlanta, GA, USA
| | - James J Lah
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
248
|
Mayorga-Weber G, Rivera FJ, Castro MA. Neuron-glia (mis)interactions in brain energy metabolism during aging. J Neurosci Res 2022; 100:835-854. [PMID: 35085408 DOI: 10.1002/jnr.25015] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/08/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023]
Abstract
Life expectancy in humans is increasing, resulting in a growing aging population, that is accompanied by an increased disposition to develop cognitive deterioration. Hypometabolism is one of the multiple factors related to inefficient brain function during aging. This review emphasizes the metabolic interactions between glial cells (astrocytes, oligodendrocytes, and microglia) and neurons, particularly, during aging. Glial cells provide support and protection to neurons allowing adequate synaptic activity. We address metabolic coupling from the expression of transporters, availability of substrates, metabolic pathways, and mitochondrial activity. In aging, the main metabolic exchange machinery is altered with inefficient levels of nutrients and detrimental mitochondrial activity that results in high reactive oxygen species levels and reduced ATP production, generating a highly inflammatory environment that favors deregulated cell death. Here, we provide an overview of the glial-to-neuron mechanisms, from the molecular components to the cell types, emphasizing aging as the crucial risk factor for developing neurodegenerative/neuroinflammatory diseases.
Collapse
Affiliation(s)
- Gonzalo Mayorga-Weber
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Francisco J Rivera
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Laboratory of Stem Cells and Neuroregeneration, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Janelia Research Campus, HHMI, Ashburn, VA, USA
| |
Collapse
|
249
|
Vespakinin-M, a natural peptide from Vespa magnifica, promotes functional recovery in stroke mice. Commun Biol 2022; 5:74. [PMID: 35058552 PMCID: PMC8776894 DOI: 10.1038/s42003-022-03024-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Acute ischemic stroke triggers complex systemic pathological responses for which the exploration of drug resources remains a challenge. Wasp venom extracted from Vespa magnifica (Smith, 1852) is most commonly used to treat rheumatoid arthritis as well as neurological disorders. Vespakinin-M (VK), a natural peptide from wasp venom, has remained largely unexplored for stroke. Herein, we first confirmed the structure, stability, toxicity and distribution of VK as well as its penetration into the blood–brain barrier. VK (150 and 300 µg/kg, i.p.) was administered to improve stroke constructed by middle cerebral artery occlusion in mice. Our results indicate that VK promote functional recovery in mice after ischemia stroke, including an improvement of neurological impairment, reduction of infarct volume, maintenance of blood-brain barrier integrity, and an obstruction of the inflammatory response and oxidative stress. In addition, VK treatment led to reduced neuroinflammation and apoptosis associated with the activation of PI3K–AKT and inhibition of IκBα–NF-κB signaling pathways. Simultaneously, we confirmed that VK can combine with bradykinin receptor 2 (B2R) as detected by molecular docking, the B2R antagonist HOE140 could counteract the neuro-protective effects of VK on stroke in mice. Overall, targeting the VK–B2R interaction can be considered as a practical strategy for stroke therapy. Zhao et al establish the structure, stability, toxicity and distribution of vespakinin-M (VK) as well as its penetration into the blood–brain barrier in mice. They go on to show that VK promotes functional recovery in mice after ischemia stroke and shed light on the potential underlying mechanisms, which together indicates the potential therapeutic value of targeting VK in stroke therapy.
Collapse
|
250
|
Lee JW, Profant M, Wang C. Metabolic Sex Dimorphism of the Brain at the Gene, Cell, and Tissue Level. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:212-220. [PMID: 35017210 DOI: 10.4049/jimmunol.2100853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/09/2021] [Indexed: 12/21/2022]
Abstract
The palpable observation in the sex bias of disease prevalence in the CNS has fascinated scientists for several generations. Brain sex dimorphism has been visualized by imaging and analytical tools at the tissue, cellular, and molecular levels. Recent work highlighted the specificity of such sex bias in the brain and its subregions, offering a unique lens through which disease pathogenesis can be investigated. The brain is the largest consumer of energy in the body and provides a unique metabolic environment for diverse lineages of cells. Immune cells are increasingly recognized as an integral part of brain physiology, and their function depends on metabolic homeostasis. This review focuses on metabolic sex dimorphism in brain tissue, resident, and infiltrating immune cells. In this context, we highlight the relevance of recent advances in metabolomics and RNA sequencing technologies at the single cell resolution and the development of novel computational approaches.
Collapse
Affiliation(s)
- Jun Won Lee
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and
| | - Martin Profant
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Chao Wang
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and .,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|