1
|
Mo H, Zhang X, Ren L. Analysis of neuroglia and immune cells in the tumor microenvironment of breast cancer brain metastasis. Cancer Biol Ther 2024; 25:2398285. [PMID: 39238191 PMCID: PMC11382727 DOI: 10.1080/15384047.2024.2398285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
Breast cancer stands as the most prevalent cancer diagnosed worldwide, often leading to brain metastasis, a challenging complication characterized by high mortality rates and a grim prognosis. Understanding the intricate mechanisms governing breast cancer brain metastasis (BCBM) remains an ongoing challenge. The unique microenvironment in the brain fosters an ideal setting for the colonization of breast cancer cells. The tumor microenvironment (TME) in brain metastases plays a pivotal role in the initiation and progression of BCBM, shaping the landscape for targeted therapeutic interventions. Current research primarily concentrates on unraveling the complexities of the TME in BCBM, with a particular emphasis on neuroglia and immune cells, such as microglia, monocyte-derived macrophages (MDMs), astrocytes and T cells. This comprehensive review delves deeply into these elements within the TME of BCBM, shedding light on their interplay, mechanisms, and potential as therapeutic targets to combat BCBM.
Collapse
Affiliation(s)
- Haixin Mo
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Liangliang Ren
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| |
Collapse
|
2
|
Chen Z, Ford KP, Islam MBAR, Wan H, Han H, Ramakrishnan A, Brown RJ, Villanueva V, Wang Y, Davis BT, Weiss C, Cui W, Gate D, Schwulst SJ. Anti-CD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8 + T-cell cytotoxicity after traumatic brain injury. J Neuroinflammation 2024; 21:267. [PMID: 39427160 PMCID: PMC11491007 DOI: 10.1186/s12974-024-03257-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people.
Collapse
Affiliation(s)
- Zhangying Chen
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Kacie P Ford
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mecca B A R Islam
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hanxiao Wan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hyebin Han
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Abhirami Ramakrishnan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ryan J Brown
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Veronica Villanueva
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yidan Wang
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Booker T Davis
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Craig Weiss
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Weiguo Cui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Steven J Schwulst
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
3
|
Zhou T, Gao Y, Wang Z, Dai C, Lei M, Liew A, Yan S, Yao Z, Hu D, Qi F. CD8 positive T-cells decrease neurogenesis and induce anxiety-like behaviour following hepatitis B vaccination. Brain Commun 2024; 6:fcae315. [PMID: 39386089 PMCID: PMC11462449 DOI: 10.1093/braincomms/fcae315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/23/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Mounting evidence indicates the involvement of peripheral immunity in the regulation of brain function, influencing aspects such as neuronal development, emotion, and cognitive abilities. Previous studies from our laboratory have revealed that neonatal hepatitis B vaccination can downregulate hippocampal neurogenesis, synaptic plasticity and spatial learning memory. In the current post-epidemic era characterized by universal vaccination, understanding the impact of acquired immunity on neuronal function and neuropsychiatric disorders, along with exploring potential underlying mechanisms, becomes imperative. We employed hepatitis B vaccine-induced CD3 positive T cells in immunodeficient mice to investigate the key mechanisms through which T cell subsets modulate hippocampal neurogenesis and anxiety-like behaviours. Our data revealed that mice receiving hepatitis B vaccine-induced T cells exhibited heightened anxiety and decreased hippocampal cell proliferation compared to those receiving phosphate-buffered saline-T cells or wild-type mice. Importantly, these changes were predominantly mediated by infiltrated CD8+ T cells into the brain, rather than CD4+ T cells. Transcriptome profiling of CD8+ T cells unveiled that C-X-C motif chemokine receptor 6 positive (CXCR6+) CD8+ T cells were recruited into the brain through microglial and astrocyte-derived C-X-C motif chemokine ligand 16 (CXCL16). This recruitment process impaired neurogenesis and induced anxiety-like behaviour via tumour necrosis factor-α-dependent mechanisms. Our findings highlight the role of glial cell derived CXCL16 in mediating the recruitment of CXCR6+CD8+ T cell subsets into the brain. This mechanism represents a potential avenue for modulating hippocampal neurogenesis and emotion-related behaviours after hepatitis B vaccination.
Collapse
Affiliation(s)
- Tuo Zhou
- Children's Health Section, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Yuxuan Gao
- Breast Disease Center, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhiling Wang
- Department of Orthopedic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Chunfang Dai
- Children's Health Section, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ming Lei
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Aubrey Liew
- Department of Immunology, Mayo Clinic, Rochester 55905, USA
| | - Sen Yan
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 519070, China
| | - Zhibin Yao
- Department of Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Dandan Hu
- Children's Health Section, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Fangfang Qi
- Department of Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
- Department of Neurology, Mayo Clinic, Rochester 55905, USA
| |
Collapse
|
4
|
Qiao Y, Mei Y, Xia M, Luo D, Gao L. The role of m6A modification in the risk prediction and Notch1 pathway of Alzheimer's disease. iScience 2024; 27:110235. [PMID: 39040060 PMCID: PMC11261416 DOI: 10.1016/j.isci.2024.110235] [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: 06/26/2023] [Revised: 03/17/2024] [Accepted: 05/19/2024] [Indexed: 07/24/2024] Open
Abstract
N6-methyladenosine (m6A) methylation and abnormal immune responses are implicated in neurodegenerative diseases, yet their relationship in Alzheimer's disease (AD) remains unclear. We obtained AD datasets from GEO databases and used AD mouse and cell models, observing abnormal expression of m6A genes in the AD group, alongside disruptions in the immune microenvironment. Key m6A genes (YTHDF2, LRPPRC, and FTO) selected by machine learning were associated with the Notch pathway, with FTO and Notch1 displaying the strongest correlation. Specifically, FTO expression decreased and m6A methylation of Notch1 increased in AD mouse and cell models. We further silenced FTO expression in HT22 cells, resulting in upregulation of the Notch1 signaling pathway. Additionally, increased Notch1 expression in dendritic cells heightened inflammatory cytokine secretion in vitro. These results suggest that reduced FTO expression may contribute to the pathogenesis of AD by activating the Notch1 pathway to interfere with the immune response.
Collapse
Affiliation(s)
- Yingdan Qiao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Yingna Mei
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Minqi Xia
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Deng Luo
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| |
Collapse
|
5
|
Panwar A, Rentsendorj A, Jhun M, Cohen RM, Cordner R, Gull N, Pechnick RN, Duvall G, Mardiros A, Golchian D, Schubloom H, Jin LW, Van Dam D, Vermeiren Y, De Reu H, De Deyn PP, Raskatov JA, Black KL, Irvin DK, Williams BA, Wheeler CJ. Antigen-specific age-related memory CD8 T cells induce and track Alzheimer's-like neurodegeneration. Proc Natl Acad Sci U S A 2024; 121:e2401420121. [PMID: 38995966 PMCID: PMC11260139 DOI: 10.1073/pnas.2401420121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/23/2024] [Indexed: 07/14/2024] Open
Abstract
Cerebral (Aβ) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aβ/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aβ/pTau, however, appears to vary depending on the animal model. Our prior work suggested that antigen-specific memory CD8 T ("hiT") cells act upstream of Aβ/pTau after brain injury. Here, we examine whether hiT cells influence sporadic AD-like pathophysiology upstream of Aβ/pTau. Examining neuropathology, gene expression, and behavior in our hiT mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. We identify an age-related factor acting upstream of Aβ/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD.
Collapse
Affiliation(s)
- Akanksha Panwar
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Altan Rentsendorj
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Michelle Jhun
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Robert M. Cohen
- Department Psychiatry & Behavioral Sciences and Neuroscience Program, Graduate Division of Biological and Biomedical Sciences (GDBBS), Emory University, Atlanta, GA30322
| | - Ryan Cordner
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
- Department Biomedical & Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Nicole Gull
- Department Biomedical & Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Robert N. Pechnick
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, CA91766
| | - Gretchen Duvall
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Armen Mardiros
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - David Golchian
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Hannah Schubloom
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Lee-Way Jin
- Department Medical Pathology and Laboratory Medicine, Laboratory Medicine, Medical Investigation of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California, Davis, Sacramento, CA95817
| | - Debby Van Dam
- Department of Biomedical Sciences, Institute Born-Bunge, Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp2610, Belgium
- Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen AB9700, Netherlands
| | - Yannick Vermeiren
- Department of Biomedical Sciences, Institute Born-Bunge, Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp2610, Belgium
- Faculty of Medicine & Health Sciences, Department of Translational Neurosciences, University of Antwerp, Antwerp2610, Belgium
- Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University & Research, Wageningen AA6700, The Netherlands
| | - Hans De Reu
- Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute, Laboratory of Experimental Hematology, University of Antwerp, Antwerp2610, Belgium
| | - Peter Paul De Deyn
- Department of Biomedical Sciences, Institute Born-Bunge, Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp2610, Belgium
- Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen AB9700, Netherlands
- Department of Neurology, Memory Clinic of Hospital Network Antwerp, Middelheim and Hoge Beuken, Antwerp BE-2660, Belgium
- Department of Chemistry & Biochemistry, University of California, Santa Cruz, CA95064
| | - Jevgenij A. Raskatov
- Department of Chemistry & Biochemistry, University of California, Santa Cruz, CA95064
| | - Keith L. Black
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Dwain K. Irvin
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
- NovAccess Global and StemVax LLC, Cleveland, OH44023
| | - Brian A. Williams
- Transcriptome Function and Technology Program, Department of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Christopher J. Wheeler
- Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA90048
- Department of Chemistry & Biochemistry, University of California, Santa Cruz, CA95064
- NovAccess Global and StemVax LLC, Cleveland, OH44023
- Society for Brain Mapping & Therapeutics, World Brain Mapping Foundation, Pacific Palisades, CA90272
- T-Neuro Pharma, Inc., Albuquerque, NM87123
| |
Collapse
|
6
|
Farris T, González-Ochoa S, Mohammed M, Rajakaruna H, Tonello J, Kanagasabai T, Korolkova O, Shimamoto A, Ivanova A, Shanker A. Loss of Mitochondrial Tusc2/Fus1 Triggers a Brain Pro-Inflammatory Microenvironment and Early Spatial Memory Impairment. Int J Mol Sci 2024; 25:7406. [PMID: 39000512 PMCID: PMC11242373 DOI: 10.3390/ijms25137406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Brain pathological changes impair cognition early in disease etiology. There is an urgent need to understand aging-linked mechanisms of early memory loss to develop therapeutic strategies and prevent the development of cognitive impairment. Tusc2 is a mitochondrial-resident protein regulating Ca2+ fluxes to and from mitochondria impacting overall health. We previously reported that Tusc2-/- female mice develop chronic inflammation and age prematurely, causing age- and sex-dependent spatial memory deficits at 5 months old. Therefore, we investigated Tusc2-dependent mechanisms of memory impairment in 4-month-old mice, comparing changes in resident and brain-infiltrating immune cells. Interestingly, Tusc2-/- female mice demonstrated a pro-inflammatory increase in astrocytes, expression of IFN-γ in CD4+ T cells and Granzyme-B in CD8+T cells. We also found fewer FOXP3+ T-regulatory cells and Ly49G+ NK and Ly49G+ NKT cells in female Tusc2-/- brains, suggesting a dampened anti-inflammatory response. Moreover, Tusc2-/- hippocampi exhibited Tusc2- and sex-specific protein changes associated with brain plasticity, including mTOR activation, and Calbindin and CamKII dysregulation affecting intracellular Ca2+ dynamics. Overall, the data suggest that dysregulation of Ca2+-dependent processes and a heightened pro-inflammatory brain microenvironment in Tusc2-/- mice could underlie cognitive impairment. Thus, strategies to modulate the mitochondrial Tusc2- and Ca2+- signaling pathways in the brain should be explored to improve cognitive health.
Collapse
Affiliation(s)
- Tonie Farris
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Salvador González-Ochoa
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Muna Mohammed
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Harshana Rajakaruna
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| | - Jane Tonello
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Olga Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Alla Ivanova
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
7
|
Liao J, Zhang Y, Tang Z, Liu P, He L. Causal relationships between peripheral immune cells and Alzheimer's disease: a two-sample Mendelian randomization study. Neurol Sci 2024; 45:3117-3124. [PMID: 38267604 DOI: 10.1007/s10072-024-07324-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024]
Abstract
OBJECTIVE Previous research suggests that peripheral immune cells may play a role in the development of Alzheimer's disease (AD). Our study aims to determine if the composition of peripheral immune cells directly contributes to the occurrence of AD. METHODS We utilized a two-sample Mendelian randomization (MR) approach to examine the association between peripheral immune cells and AD.The primary analysis method used was the inverse variance weighted (IVW) method, and we also conducted analyses using MR Egger, weighted median, simple mode, and weighted mode methods to ensure the accuracy of the results.Heterogeneity and horizontal pleiotropy were evaluated using Cochran's Q statistics and the MR Egger intercept, respectively. RESULTS The study found a significant correlation between increased IgD + CD24- AC cells (Odds Ratio [OR] = 1.03, 95% Confidence Interval [CI] = 1.01-1.06, P = 0.0172), increased CD4 + %leukocyte (OR = 1.08, 95% CI = 1.02-1.14, P = 0.0086), and increased CD4 + CD8dim AC cells (OR = 1.06, 95% CI = 1.01-1.11, P = 0.0218), with an increased susceptibility to AD. Conversely, an increase in EM DN (CD4-CD8-) %T cells (OR = 0.95, 95% CI = 0.92-0.99, P = 0.0164) and an increase in DN (CD4-CD8-) AC cells (OR = 0.93, 95% CI = 0.88-0.99, P = 0.0145) were associated with a protective effect against AD. CONCLUSION Our findings establish a causal link between peripheral immune cells and AD. This study is the first to examine the relationship between peripheral immune cells and AD using MR, offering valuable insights for early diagnosis and treatment decisions.
Collapse
Affiliation(s)
- Jing Liao
- Ruikang Hospital, Affiliated to Guangxi University of Chinese Medicine, 10 Huadong Road, Xingning District, Nanning City, Guangxi, 53000, China.
| | - Yongquan Zhang
- Ruikang Hospital, Affiliated to Guangxi University of Chinese Medicine, 10 Huadong Road, Xingning District, Nanning City, Guangxi, 53000, China
| | - Zhanhong Tang
- First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning City, Guangxi, 530021, China
| | - Pinjing Liu
- Ruikang Hospital, Affiliated to Guangxi University of Chinese Medicine, 10 Huadong Road, Xingning District, Nanning City, Guangxi, 53000, China
| | - Luoyi He
- Ruikang Hospital, Affiliated to Guangxi University of Chinese Medicine, 10 Huadong Road, Xingning District, Nanning City, Guangxi, 53000, China
| |
Collapse
|
8
|
Chauquet S, Willis EF, Grice L, Harley SBR, Powell JE, Wray NR, Nguyen Q, Ruitenberg MJ, Shah S, Vukovic J. Exercise rejuvenates microglia and reverses T cell accumulation in the aged female mouse brain. Aging Cell 2024; 23:e14172. [PMID: 38747044 PMCID: PMC11258432 DOI: 10.1111/acel.14172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 02/16/2024] [Accepted: 04/01/2024] [Indexed: 07/21/2024] Open
Abstract
Slowing and/or reversing brain ageing may alleviate cognitive impairments. Previous studies have found that exercise may mitigate cognitive decline, but the mechanisms underlying this remain largely unclear. Here we provide unbiased analyses of single-cell RNA sequencing data, showing the impacts of exercise and ageing on specific cell types in the mouse hippocampus. We demonstrate that exercise has a profound and selective effect on aged microglia, reverting their gene expression signature to that of young microglia. Pharmacologic depletion of microglia further demonstrated that these cells are required for the stimulatory effects of exercise on hippocampal neurogenesis but not cognition. Strikingly, allowing 18-month-old mice access to a running wheel did by and large also prevent and/or revert T cell presence in the ageing hippocampus. Taken together, our data highlight the profound impact of exercise in rejuvenating aged microglia, associated pro-neurogenic effects and on peripheral immune cell presence in the ageing female mouse brain.
Collapse
Affiliation(s)
- Solal Chauquet
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Emily F. Willis
- School of Biomedical Sciences, Faculty of MedicineThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Laura Grice
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Samuel B. R. Harley
- Queensland Brain Institute, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Joseph E. Powell
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Naomi R. Wray
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
- Department of PsychiatryUniversity of OxfordOxfordUK
- Oxford Big Data Institute, Li Ka Shing Centre for Health Information and DiscoveryUniversity of OxfordOxfordUK
| | - Quan Nguyen
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Marc J. Ruitenberg
- School of Biomedical Sciences, Faculty of MedicineThe University of QueenslandSaint LuciaQueenslandAustralia
| | - Sonia Shah
- Institute for Molecular Bioscience, the University of QueenslandSaint LuciaQueenslandAustralia
| | - Jana Vukovic
- School of Biomedical Sciences, Faculty of MedicineThe University of QueenslandSaint LuciaQueenslandAustralia
- Queensland Brain Institute, the University of QueenslandSaint LuciaQueenslandAustralia
| |
Collapse
|
9
|
Chen Z, Ford KP, Islam MBAR, Wan H, Han H, Ramakrishnan A, Brown RJ, Villanueva V, Wang Y, Davis BT, Weiss C, Cui W, Gate D, Schwulst SJ. antiCD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8+ T-cell cytotoxicity after traumatic brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.596673. [PMID: 38948775 PMCID: PMC11212861 DOI: 10.1101/2024.06.17.596673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people. Graphic abstract Aged brains after TBI comprise two pools of CD8 + T cells . The aged brain has long been resided by a population of CD8 + T cells that's exhaustive and dysfunctional. Post TBI, due to BBB impairment, functional CD8 + T cells primarily migrate into the brain parenchyma. Aged, injury-associated microglia with upregulated MHC class I molecules can present neoantigens such as neuronal and/or myelin debris in the injured brains to functional CD8+ T, resulting in downstream CD8+ T cell cytotoxicity. aCD49d Ab treatment exerts its function by blocking the migration of functional effector CD8 + T cell population, leading to less cytotoxicity and resulting in improved TBI outcomes in aged mice.
Collapse
|
10
|
Li C, Stebbins RC, Noppert GA, Carney CX, Liu C, Sapp ARM, Watson EJ, Aiello AE. Peripheral immune function and Alzheimer's disease: a living systematic review and critical appraisal. Mol Psychiatry 2024; 29:1895-1905. [PMID: 38102484 PMCID: PMC11483233 DOI: 10.1038/s41380-023-02355-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND A growing body of literature examines the relationship between peripheral immune function and Alzheimer's Disease (AD) in human populations. Our living systematic review summarizes the characteristics and findings of these studies, appraises their quality, and formulates recommendations for future research. METHODS We searched the electronic databases PubMed, PsycINFO, and Web of Science, and reviewed references of previous reviews and meta-analyses to identify human studies examining the relationship between any peripheral immune biomarkers and AD up to September 7th, 2023. We examined patterns of reported statistical associations (positive, negative, and null) between each biomarker and AD across studies. Evidence for each biomarker was categorized into four groups based on the proportion of studies reporting different associations: corroborating a positive association with AD, a negative association, a null association, and presenting contradictory findings. A modified Newcastle-Ottawa scale (NOS) was employed to assess the quality of the included studies. FINDINGS In total, 286 studies were included in this review. The majority were cross-sectional (n = 245, 85.7%) and hospital-based (n = 248, 86.7%), examining relationships between 187 different peripheral immune biomarkers and AD. Cytokines were the most frequently studied group of peripheral immune biomarkers. Evidence supported a positive association with AD for six biomarkers, including IL-6, IL-1β, IFN-γ, ACT, IL-18, and IL-12, and a negative association for two biomarkers, including lymphocytes and IL-6R. Only a small proportion of included studies (n = 22, 7.7%) were deemed to be of high quality based on quality assessment. INTERPRETATION Existing research on peripheral immune function and AD exhibits substantial methodological variations and limitations, with a notable lack of longitudinal, population-based studies investigating a broad range of biomarkers with prospective AD outcomes. The extent and manner in which peripheral immune function can contribute to AD pathophysiology remain open questions. Given the biomarkers that we identified to be associated with AD, we posit that targeting peripheral immune dysregulation may present a promising intervention point to reduce the burden of AD.
Collapse
Affiliation(s)
- Chihua Li
- Social Environment and Health Program, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology, School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| | - Rebecca C Stebbins
- Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York City, NY, USA
| | - Grace A Noppert
- Social Environment and Health Program, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Constanza X Carney
- Department of Epidemiology, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Chunyu Liu
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Ashley R M Sapp
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elijah J Watson
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Allison E Aiello
- Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York City, NY, USA
- Department of Epidemiology, Mailman School of Public, Columbia University, New York City, NY, USA
| |
Collapse
|
11
|
Sun Y, Jiang M, Long X, Miao Y, Du H, Zhang T, Ma X, Zhang Y, Meng H. Transcriptomic Analysis of Lipid Metabolism Genes in Alzheimer's Disease: Highlighting Pathological Outcomes and Compartmentalized Immune Status. J Mol Neurosci 2024; 74:55. [PMID: 38776015 DOI: 10.1007/s12031-024-02225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 07/20/2024]
Abstract
The dysregulation of lipid metabolism has been strongly associated with Alzheimer's disease (AD) and has intricate connections with various aspects of disease progression, such as amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. Here, a comprehensive bioinformatic assessment was conducted on lipid metabolism genes in the brains and peripheral blood of AD-derived transcriptome datasets, characterizing the correlation between differentially expressed genes (DEGs) of lipid metabolism and disease pathologies, as well as immune cell preferences. Through the application of weighted gene co-expression network analysis (WGCNA), modules eigengenes related to lipid metabolism were pinpointed, and the examination of their molecular functions within biological processes, molecular pathways, and their associations with pathological phenotypes and molecular networks has been characterized. Analysis of biological networks indicates notable discrepancies in the expression patterns of the DEGs between neuronal and immune cells, as well as variations in cell type enrichments within both brain tissue and peripheral blood. Additionally, drugs targeting the DEGs from central and peripheral and a diagnostic model for hub genes from the blood were retrieved and assessed, some of which were shown to be useful for therapeutic and diagnostic. These results revealed the distinctive pattern of transcriptionally abnormal lipid metabolism in central, peripheral, and immune cell activation, providing valuable insight into lipid metabolism for diagnosing and guiding more effective treatment for AD.
Collapse
Affiliation(s)
- Yue Sun
- Department of Neurology, Shanghai East Hospital of Tongji University School of Medicine, Shanghai, 20092, People's Republic of China
| | - Mengni Jiang
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Xiang Long
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Yongzhen Miao
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Huanhuan Du
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Ting Zhang
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China
| | - Xuejun Ma
- School of Mathematical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yue Zhang
- Department of Neurology, Shanghai East Hospital of Tongji University School of Medicine, Shanghai, 20092, People's Republic of China.
| | - Hongrui Meng
- Institute of Neuroscience, Soochow University, Suzhou, 215123, People's Republic of China.
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.
| |
Collapse
|
12
|
Kwok AJ, Lu J, Huang J, Ip BY, Mok VCT, Lai HM, Ko H. High-resolution omics of vascular ageing and inflammatory pathways in neurodegeneration. Semin Cell Dev Biol 2024; 155:30-49. [PMID: 37380595 DOI: 10.1016/j.semcdb.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/07/2023] [Indexed: 06/30/2023]
Abstract
High-resolution omics, particularly single-cell and spatial transcriptomic profiling, are rapidly enhancing our comprehension of the normal molecular diversity of gliovascular cells, as well as their age-related changes that contribute to neurodegeneration. With more omic profiling studies being conducted, it is becoming increasingly essential to synthesise valuable information from the rapidly accumulating findings. In this review, we present an overview of the molecular features of neurovascular and glial cells that have been recently discovered through omic profiling, with a focus on those that have potentially significant functional implications and/or show cross-species differences between human and mouse, and that are linked to vascular deficits and inflammatory pathways in ageing and neurodegenerative disorders. Additionally, we highlight the translational applications of omic profiling, and discuss omic-based strategies to accelerate biomarker discovery and facilitate disease course-modifying therapeutics development for neurodegenerative conditions.
Collapse
Affiliation(s)
- Andrew J Kwok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jianning Lu
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Junzhe Huang
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bonaventure Y Ip
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent C T Mok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hei Ming Lai
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
13
|
Hu D, Weiner HL. Unraveling the dual nature of brain CD8 + T cells in Alzheimer's disease. Mol Neurodegener 2024; 19:16. [PMID: 38355649 PMCID: PMC10865558 DOI: 10.1186/s13024-024-00706-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Affiliation(s)
- Dan Hu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 02115, Boston, MA, USA.
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 02115, Boston, MA, USA.
| |
Collapse
|
14
|
Panwar A, Rentsendorj A, Jhun M, Cohen RM, Cordner R, Gull N, Pechnick RN, Duvall G, Mardiros A, Golchian D, Schubloom H, Jin LW, Van Dam D, Vermeiren Y, De Reu H, De Deyn PP, Raskatov JA, Black KL, Irvin DK, Williams BA, Wheeler CJ. Antigen-specific age-related memory CD8 T cells induce and track Alzheimer's-like neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576704. [PMID: 38328072 PMCID: PMC10849535 DOI: 10.1101/2024.01.22.576704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Cerebral (Aβ) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aβ/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aβ/fibrillar pTau, however, appears to vary depending on the animal model used. Our prior work suggested that antigen-specific memory CD8 T (" hi T") cells act upstream of Aβ/pTau after brain injury. Here we examine whether hi T cells influence sporadic AD-like pathophysiology upstream of Aβ/pTau. Examining neuropathology, gene expression, and behavior in our hi T mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. Our work is the first to identify an age-related factor acting upstream of Aβ/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD. Significance Statement This study changes our view of Alzheimer's Disease (AD) initiation and progression. Mutations promoting cerebral beta-amyloid (Aβ) deposition guarantee rare genetic forms of AD. Thus, the prevailing hypothesis has been that Aβ is central to initiation and progression of all AD, despite contrary animal and patient evidence. We show that age-related T cells generate neurodegeneration with compelling features of AD in mice, with distinct T cell functions required for pathological initiation and neurodegenerative progression. Knowledge from these mice was applied to successfully predict previously unknown features of human AD and generate novel tools for its clinical management.
Collapse
|
15
|
Chen Y, Li Y, Fan Y, Chen S, Chen L, Chen Y, Chen Y. Gut microbiota-driven metabolic alterations reveal gut-brain communication in Alzheimer's disease model mice. Gut Microbes 2024; 16:2302310. [PMID: 38261437 PMCID: PMC10807476 DOI: 10.1080/19490976.2024.2302310] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/03/2024] [Indexed: 01/25/2024] Open
Abstract
The gut microbiota (GM) and its metabolites affect the host nervous system and are involved in the pathogeneses of various neurological diseases. However, the specific GM alterations under pathogenetic pressure and their contributions to the "microbiota - metabolite - brain axis" in Alzheimer's disease (AD) remain unclear. Here, we investigated the GM and the fecal, serum, cortical metabolomes in APP/PS1 and wild-type (WT) mice, revealing distinct hub bacteria in AD mice within scale-free GM networks shared by both groups. Moreover, we identified diverse peripheral - central metabolic landscapes between AD and WT mice that featured bile acids (e.g. deoxycholic and isodeoxycholic acid) and unsaturated fatty acids (e.g. 11Z-eicosenoic and palmitoleic acid). Machine-learning models revealed the relationships between the differential/hub bacteria and these metabolic signatures from the periphery to the brain. Notably, AD-enriched Dubosiella affected AD occurrence via cortical palmitoleic acid and vice versa. Considering the transgenic background of the AD mice, we propose that Dubosiella enrichment impedes AD progression via the synthesis of palmitoleic acid, which has protective properties against inflammation and metabolic disorders. We identified another association involving fecal deoxycholic acid-mediated interactions between the AD hub bacteria Erysipelatoclostridium and AD occurrence, which was corroborated by the correlation between deoxycholate levels and cognitive scores in humans. Overall, this study elucidated the GM network alterations, contributions of the GM to peripheral - central metabolic landscapes, and mediatory roles of metabolites between the GM and AD occurrence, thus revealing the critical roles of bacteria in AD pathogenesis and gut - brain communications under pathogenetic pressure.
Collapse
Affiliation(s)
- Yijing Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yinhu Li
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yingying Fan
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Shuai Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Li Chen
- Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
16
|
de Fàbregues O, Sellés M, Ramos-Vicente D, Roch G, Vila M, Bové J. Relevance of tissue-resident memory CD8 T cells in the onset of Parkinson's disease and examination of its possible etiologies: infectious or autoimmune? Neurobiol Dis 2023; 187:106308. [PMID: 37741513 DOI: 10.1016/j.nbd.2023.106308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/05/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023] Open
Abstract
Tissue-resident memory CD8 T cells are responsible for local immune surveillance in different tissues, including the brain. They constitute the first line of defense against pathogens and cancer cells and play a role in autoimmunity. A recently published study demonstrated that CD8 T cells with markers of residency containing distinct granzymes and interferon-γ infiltrate the parenchyma of the substantia nigra and contact dopaminergic neurons in an early premotor stage of Parkinson's disease. This infiltration precedes α-synuclein aggregation and neuronal loss in the substantia nigra, suggesting a relevant role for CD8 T cells in the onset of the disease. To date, the nature of the antigen that initiates the adaptive immune response remains unknown. This review will discuss the role of tissue-resident memory CD8 T cells in brain immune homeostasis and in the onset of Parkinson's disease and other neurological diseases. We also discuss how aging and genetic factors can affect the CD8 T cell immune response and how animal models can be misleading when studying human-related immune response. Finally, we speculate about a possible infectious or autoimmune origin of Parkinson's disease.
Collapse
Affiliation(s)
- Oriol de Fàbregues
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain; Movement Disorders Unit, Neurology Department, Vall d'Hebron University Hospital
| | - Maria Sellés
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - David Ramos-Vicente
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Gerard Roch
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain; Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Catalonia, Spain; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - Jordi Bové
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain.
| |
Collapse
|
17
|
Li X, Wang H, Zhang Q, Sun X, Zhang M, Wang G. Inhibition of adult hippocampal neurogenesis induced by postoperative CD8 + T-cell infiltration is associated with cognitive decline later following surgery in adult mice. J Neuroinflammation 2023; 20:227. [PMID: 37798730 PMCID: PMC10557222 DOI: 10.1186/s12974-023-02910-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Some patients show persistent cognitive decline for weeks, months or even years after surgery, which seriously affects their long-term prognosis and quality of life. However, most previous basic studies have focused mainly on the mechanisms of early postoperative cognitive decline, whereas cognitive decline in the longer term after surgery is less well-understood. The subgranular zone of the dentate gyrus exhibits life-long neurogenesis, supporting hippocampus-dependent learning and memory. MAIN TEXT The aim of this study was to investigate whether adult hippocampal neurogenesis (AHN) involves in cognitive decline later following surgery and to further explore the roles of CD8 + T lymphocytes infiltrating the hippocampal parenchyma after surgery in this pathological process. Cognitive function was examined in adult mice that underwent laparotomy combined with partial hepatectomy, and the results showed that cognitive decline persisted in mice who underwent surgery during the first postoperative month, even though there was a trend toward continuous improvement over time. Significantly decreased numbers of DCX + cells, BrdU + cells, and BrdU + /DCX + cells were observed on day 8 after surgery, and a significantly decreased number of NeuN + /BrdU + cells was observed on day 28 after surgery, which indicated inhibition of AHN. After surgery, T lymphocytes, the majority of which were CD8 + T cells, infiltrated the hippocampus and secreted Interferon-γ (IFN-γ). Depletion of CD8 + T cells could inhibit the increase of IFN-γ synthesis, improve hippocampal neurogenesis, and improve postoperative cognitive function. Hippocampal microinjection of IFN-γ neutralizing antibody or adeno-associated virus to knock down IFN-γ receptor 1 (IFNGR1) could also partially attenuate the inhibition of AHN and improve postoperative cognitive function. CONCLUSIONS These results demonstrate that postoperative infiltration of CD8 + T cells into the hippocampus and subsequent secretion of IFN-γ contribute to the inhibition of AHN and cognitive decline later following surgery.
Collapse
Affiliation(s)
- Xiaowei Li
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hong Wang
- Department of Nephrology, Tai' an Central Hospital, Taian, 271000, Shandong, China
| | - Qidi Zhang
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaobin Sun
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
18
|
Jin J, Guang M, Li S, Liu Y, Zhang L, Zhang B, Cheng M, Schmalz G, Huang X. Immune-related signature of periodontitis and Alzheimer's disease linkage. Front Genet 2023; 14:1230245. [PMID: 37849501 PMCID: PMC10577303 DOI: 10.3389/fgene.2023.1230245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023] Open
Abstract
Background: Periodontits (PD) and Alzheimer's disease (AD) are both associated with ageing and clinical studies increasingly evidence their association. However, specific mechanisms underlying this association remain undeciphered, and immune-related processes are purported to play a signifcant role. The accrual of publicly available transcriptomic datasets permits secondary analysis and the application of data-mining and bioinformatic tools for biological discovery. Aim: The present study aimed to leverage publicly available transcriptomic datasets and databases, and apply a series of bioinformatic analysis to identify a robust signature of immune-related signature of PD and AD linkage. Methods: We downloaded gene-expresssion data pertaining PD and AD and identified crosstalk genes. We constructed a protein-protein network analysis, applied immune cell enrichment analysis, and predicted crosstalk immune-related genes and infiltrating immune cells. Next, we applied consisent cluster analysis and performed immune cell bias analysis, followed by LASSO regression to select biomarker immune-related genes. Results: The results showed a 3 gene set comprising of DUSP14, F13A1 and SELE as a robust immune-related signature. Macrophages M2 and NKT, B-cells, CD4+ memory T-cells and CD8+ naive T-cells emerged as key immune cells linking PD with AD. Conclusion: Candidate immune-related biomarker genes and immune cells central to the assocation of PD with AD were identified, and merit investigation in experimental and clinical research.
Collapse
Affiliation(s)
- Jieqi Jin
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mengkai Guang
- Department of Stomatology, China-Japan Friendship Hospital, Beijing, China
| | - Simin Li
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yong Liu
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liwei Zhang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bo Zhang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Menglin Cheng
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Gerhard Schmalz
- Department of Cariology, Endodontology and Periodontology, Leipzig University, Leipzig, Germany
| | - Xiaofeng Huang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Berger-Sieczkowski E, Endmayr V, Haider C, Ricken G, Jauk P, Macher S, Pirker W, Högl B, Heidbreder A, Schnider P, Bradley-Zechmeister E, Mariotto S, Koneczny I, Reinecke R, Kasprian G, Weber C, Bergmann M, Milenkovic I, Berger T, Gaig C, Sabater L, Graus F, Gelpi E, Höftberger R. Analysis of inflammatory markers and tau deposits in an autopsy series of nine patients with anti-IgLON5 disease. Acta Neuropathol 2023; 146:631-645. [PMID: 37646790 PMCID: PMC10499680 DOI: 10.1007/s00401-023-02625-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023]
Abstract
Anti-IgLON5 disease is a rare neurological, probably autoimmune, disorder associated in many cases with a specific tauopathy. Only a few post-mortem neuropathological studies have been reported so far. Little is known about the pathogenic mechanisms that result in neurodegeneration. We investigated the neuropathology of anti-IgLON5 disease and characterized cellular and humoral inflammation. We included nine cases (six of them previously published). Median age of patients was 71 years (53-82 years), the median disease duration was 6 years (0.5-13 years), and the female to male ratio was 5:4. Six cases with a median disease duration of 9 years presented a prominent tauopathy. Five of them had a classical anti-IgLON5-related brainstem tauopathy and another presented a prominent neuronal and glial 4-repeat tauopathy, consistent with progressive supranuclear palsy (PSP). Three cases with short disease duration (median 1.25 years) only showed a primary age-related neurofibrillary pathology. Inflammatory infiltrates of T and B cells were mild to moderate and did not significantly differ between anti-IgLON5 disease cases with or without tauopathy. In contrast, we found an extensive neuropil deposition of IgG4 in the tegmentum of the brainstem, olivary nucleus, and cerebellar cortex that was most prominent in two patients with short disease duration without the typical IgLON5-related tauopathy. The IgG4 deposits were particularly prominent in the cerebellar cortex and in these regions accompanied by mild IgG1 deposits. Activated complement deposition (C9neo) was absent. Our study indicates that IgLON5-related tau pathology occurs in later disease stages and may also present a PSP-phenotype with exclusively 4-repeat neuronal and glial tau pathology. The prominent deposition of anti-IgLON5 IgG4 at predilection sites for tau pathology suggests that anti-IgLON5 antibodies precede the tau pathology. Early start of immunotherapy might prevent irreversible neuronal damage and progression of the disease, at least in a subgroup of patients.
Collapse
Affiliation(s)
- Evelyn Berger-Sieczkowski
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Verena Endmayr
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Carmen Haider
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Gerda Ricken
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Philipp Jauk
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Stefan Macher
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Walter Pirker
- Department of Neurology, Klinik Ottakring, Vienna, Austria
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Heidbreder
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Schnider
- Department of Neurology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | | | - Sara Mariotto
- Neurology Unit, Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
| | - Inga Koneczny
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Raphael Reinecke
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Gregor Kasprian
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Corinna Weber
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Melanie Bergmann
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ivan Milenkovic
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Carles Gaig
- Neurology Service, Hospital Clínic of Barcelona, Barcelona, Spain
- Neuroimmunology Laboratory-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lidia Sabater
- Neuroimmunology Laboratory-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francesc Graus
- Neuroimmunology Laboratory-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ellen Gelpi
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Neurological Tissue Bank of the Biobanc-Hospital Clinic-IDIBAPS, Barcelona, Spain.
| | - Romana Höftberger
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
20
|
Affiliation(s)
- Katie L Reagin
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Kristen E Funk
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA.
| |
Collapse
|
21
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley KB, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. GeroScience 2023; 45:3019-3043. [PMID: 37393197 PMCID: PMC10643718 DOI: 10.1007/s11357-023-00859-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023] Open
Abstract
Major histocompatibility complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses, but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here, we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating ribosome affinity purification-qPCR analysis of 3-6- and 18-22-month-old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m, H2-D1, H2-K1, H2-M3, H2-Q6, and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I-binding leukocyte immunoglobulin-like (Lilrs) and paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell -autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A, suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M Kellogg
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Adeline H Machalinski
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Hunter L Porter
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Harris E Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla B Tooley
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Sarah R Ocañas
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA.
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
22
|
Su W, Saravia J, Risch I, Rankin S, Guy C, Chapman NM, Shi H, Sun Y, Kc A, Li W, Huang H, Lim SA, Hu H, Wang Y, Liu D, Jiao Y, Chen PC, Soliman H, Yan KK, Zhang J, Vogel P, Liu X, Serrano GE, Beach TG, Yu J, Peng J, Chi H. CXCR6 orchestrates brain CD8 + T cell residency and limits mouse Alzheimer's disease pathology. Nat Immunol 2023; 24:1735-1747. [PMID: 37679549 PMCID: PMC11102766 DOI: 10.1038/s41590-023-01604-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), are characterized by innate immune-mediated inflammation, but functional and mechanistic effects of the adaptive immune system remain unclear. Here we identify brain-resident CD8+ T cells that coexpress CXCR6 and PD-1 and are in proximity to plaque-associated microglia in human and mouse AD brains. We also establish that CD8+ T cells restrict AD pathologies, including β-amyloid deposition and cognitive decline. Ligand-receptor interaction analysis identifies CXCL16-CXCR6 intercellular communication between microglia and CD8+ T cells. Further, Cxcr6 deficiency impairs accumulation, tissue residency programming and clonal expansion of brain PD-1+CD8+ T cells. Ablation of Cxcr6 or CD8+ T cells ultimately increases proinflammatory cytokine production from microglia, with CXCR6 orchestrating brain CD8+ T cell-microglia colocalization. Collectively, our study reveals protective roles for brain CD8+ T cells and CXCR6 in mouse AD pathogenesis and highlights that microenvironment-specific, intercellular communication orchestrates tissue homeostasis and protection from neuroinflammation.
Collapse
Affiliation(s)
- Wei Su
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jordy Saravia
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Isabel Risch
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sherri Rankin
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yu Sun
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Anil Kc
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wei Li
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongling Huang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Seon Ah Lim
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Haoran Hu
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yan Wang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Danting Liu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yun Jiao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hadeer Soliman
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Koon-Kiu Yan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jonathan Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xueyan Liu
- Department of Mathematics, University of New Orleans, New Orleans, LA, USA
| | | | | | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of 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
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
23
|
Sutter PA, Ménoret A, Jellison ER, Nicaise AM, Bradbury AM, Vella AT, Bongarzone ER, Crocker SJ. CD8+ T cell depletion prevents neuropathology in a mouse model of globoid cell leukodystrophy. J Exp Med 2023; 220:e20221862. [PMID: 37310382 PMCID: PMC10266545 DOI: 10.1084/jem.20221862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/10/2023] [Accepted: 05/26/2023] [Indexed: 06/14/2023] Open
Abstract
Globoid cell leukodystrophy (GLD) or Krabbe's disease is a fatal genetic demyelinating disease of the central nervous system caused by loss-of-function mutations in the galactosylceramidase (galc) gene. While the metabolic basis for disease is known, the understanding of how this results in neuropathology is not well understood. Herein, we report that the rapid and protracted elevation of CD8+ cytotoxic T lymphocytes occurs coincident with clinical disease in a mouse model of GLD. Administration of a function-blocking antibody against CD8α effectively prevented disease onset, reduced morbidity and mortality, and prevented CNS demyelination in mice. These data indicate that subsequent to the genetic cause of disease, neuropathology is driven by pathogenic CD8+ T cells, thus offering novel therapeutic potential for treatment of GLD.
Collapse
Affiliation(s)
- Pearl A. Sutter
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Antoine Ménoret
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Evan R. Jellison
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Alexandra M. Nicaise
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Clinical Neuroscience and National Institute for Health Research Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Allison M. Bradbury
- Department of Pediatrics, Nationwide Children's Hospital, Ohio State University, Columbus, OH, USA
| | - Anthony T. Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Ernesto R. Bongarzone
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Stephen J. Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
24
|
Yang H, Qin Q, Wang M, Yin Y, Li R, Tang Y. Crosstalk between peripheral immunity and central nervous system in Alzheimer's disease. Cell Immunol 2023; 391-392:104743. [PMID: 37451918 DOI: 10.1016/j.cellimm.2023.104743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
The significance of peripheral immunity in the pathogenesis and progression of Alzheimer's diseases (AD) has been recognized. Brain-infiltrated peripheral immune components transporting across the blood-brain barrier (BBB) may reshape the central immune environment. However, mechanisms of how these components open the BBB for AD occurrence and development and correlations between peripheral and central immunity have not been fully explored. Herein, we formulate a hypothesis whereby peripheral immunity as a critical factor allows AD to progress. Peripheral central immune cell crosstalk is associated with early AD pathology and related risk factors. The damaged BBB permits peripheral immune cells to enter the central immune system to deprive its immune privilege promoting the progression toward developing AD. This review summarizes the influences of risk factors on peripheral immunity, alongside their functions, highlighting the concept of peripheral and central immunity as an integrated system in AD pathogenesis, which has received scant attention before.
Collapse
Affiliation(s)
- Hanchen Yang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qi Qin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Meng Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yunsi Yin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ruiyang Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.
| |
Collapse
|
25
|
Abdelmoaty MM, Yeapuri P, Machhi J, Lu Y, Namminga KL, Kadry R, Lu E, Bhattarai S, Mosley RL, Gendelman HE. Immune senescence in aged APP/PS1 mice. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:317-330. [PMID: 38023614 PMCID: PMC10659760 DOI: 10.1515/nipt-2023-0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 12/01/2023]
Abstract
Objectives To evaluate the linkage between age and deficits in innate and adaptive immunity which heralds both Alzheimer's disease (AD) onset and progression. The pathobiological events which underlie and tie these outcomes remain not fully understood. Methods To investigate age-dependent immunity in AD, we evaluated innate and adaptive immunity in coordinate studies of regulatory T cell (Treg) function, T cell frequencies, and microglial integrity. These were assessed in blood, peripheral lymphoid tissues, and the hippocampus of transgenic (Tg) amyloid precursor protein/presenilin 1 (APP/PS1) against non-Tg mice. Additionally, immune arrays of hippocampal tissue were performed at 4, 6, 12, and 20 months of age. Results APP/PS1 mice showed progressive impairment of Treg immunosuppressive function with age. There was partial restoration of Treg function in 20-month-old mice. Ingenuity pathway analyses of hippocampal tissues were enriched in inflammatory, oxidative, and cellular activation pathways that paralleled advancing age and AD-pathobiology. Operative genes in those pathways included, but were not limited to triggering receptor on myeloid cells 1 (TREM1), T helper type 1 (Th1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathways. Interleukin-17 (IL-17), nitric oxide, acute phase, and T cell receptor signaling pathways were also perturbed. Significant inflammation was observed at 6- and 12-months. However, at 20-months, age associated partial restoration of Treg function reduced inflammatory phenotype. Conclusions Impaired Treg function, inflammation and oxidative stress were associated with AD pathology. Age associated partial restoration of Treg function in old mice reduced the hippocampal inflammatory phenotype. Restoring Treg suppressive function can be a therapeutic modality for AD.
Collapse
Affiliation(s)
- Mai M. Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
26
|
Abbaoui A, Fatoba O, Yamashita T. Meningeal T cells function in the central nervous system homeostasis and neurodegenerative diseases. Front Cell Neurosci 2023; 17:1181071. [PMID: 37608988 PMCID: PMC10440440 DOI: 10.3389/fncel.2023.1181071] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/25/2023] [Indexed: 08/24/2023] Open
Abstract
Recently, a rising interest is given to neuroimmune communication in physiological and neuropathological conditions. Meningeal immunity is a complex immune environment housing different types of immune cells. Here, we focus on meningeal T cells, possibly the most explored aspect of neuro-immune cell interactions. Emerging data have shown that meningeal T cells play a crucial role in the pathogenesis of several neurodegenerative disorders, including multiple sclerosis, Alzheimer's, Parkinson's, and Huntington's diseases. This review highlights how meningeal T cells may contribute to immune surveillance of the central nervous system (CNS) and regulate neurobehavioral functions through the secretion of cytokines. Overall, this review assesses the recent knowledge of meningeal T cells and their effects on CNS functioning in both health and disease conditions and the underlying mechanisms.
Collapse
Affiliation(s)
- Abdellatif Abbaoui
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
- World Premier International (WPI)-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Oluwaseun Fatoba
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
- World Premier International (WPI)-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
- World Premier International (WPI)-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
27
|
Ayasoufi K, Wolf DM, Namen SL, Jin F, Tritz ZP, Pfaller CK, Zheng J, Goddery EN, Fain CE, Gulbicki LR, Borchers AL, Reesman RA, Yokanovich LT, Maynes MA, Bamkole MA, Khadka RH, Hansen MJ, Wu LJ, Johnson AJ. Brain resident memory T cells rapidly expand and initiate neuroinflammatory responses following CNS viral infection. Brain Behav Immun 2023; 112:51-76. [PMID: 37236326 PMCID: PMC10527492 DOI: 10.1016/j.bbi.2023.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/25/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
The contribution of circulating verses tissue resident memory T cells (TRMs) to clinical neuropathology is an enduring question due to a lack of mechanistic insights. The prevailing view is TRMs are protective against pathogens in the brain. However, the extent to which antigen-specific TRMs induce neuropathology upon reactivation is understudied. Using the described phenotype of TRMs, we found that brains of naïve mice harbor populations of CD69+ CD103- T cells. Notably, numbers of CD69+ CD103- TRMs rapidly increase following neurological insults of various origins. This TRM expansion precedes infiltration of virus antigen-specific CD8 T cells and is due to proliferation of T cells within the brain. We next evaluated the capacity of antigen-specific TRMs in the brain to induce significant neuroinflammation post virus clearance, including infiltration of inflammatory myeloid cells, activation of T cells in the brain, microglial activation, and significant blood brain barrier disruption. These neuroinflammatory events were induced by TRMs, as depletion of peripheral T cells or blocking T cell trafficking using FTY720 did not change the neuroinflammatory course. Depletion of all CD8 T cells, however, completely abrogated the neuroinflammatory response. Reactivation of antigen-specific TRMs in the brain also induced profound lymphopenia within the blood compartment. We have therefore determined that antigen-specific TRMs can induce significant neuroinflammation, neuropathology, and peripheral immunosuppression. The use of cognate antigen to reactivate CD8 TRMs enables us to isolate the neuropathologic effects induced by this cell type independently of other branches of immunological memory, differentiating this work from studies employing whole pathogen re-challenge. This study also demonstrates the capacity for CD8 TRMs to contribute to pathology associated with neurodegenerative disorders and long-term complications associated with viral infections. Understanding functions of brain TRMs is crucial in investigating their role in neurodegenerative disorders including MS, CNS cancers, and long-term complications associated with viral infections including COVID-19.
Collapse
Affiliation(s)
| | - Delaney M Wolf
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Shelby L Namen
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Zachariah P Tritz
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Christian K Pfaller
- Mayo Clinic Department of Molecular Medicine, Rochester, MN, United States; Paul-Ehrlich-Institut, Langen, Germany
| | - Jiaying Zheng
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Emma N Goddery
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Cori E Fain
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | | | - Anna L Borchers
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | | | - Lila T Yokanovich
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Mark A Maynes
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Michael A Bamkole
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Roman H Khadka
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Michael J Hansen
- Mayo Clinic Department of Immunology, Rochester, MN, United States
| | - Long-Jun Wu
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN, United States; Mayo Clinic Department of Molecular Medicine, Rochester, MN, United States; Mayo Clinic Department of Neurology, Rochester, MN, United States.
| |
Collapse
|
28
|
Terrabuio E, Zenaro E, Constantin G. The role of the CD8+ T cell compartment in ageing and neurodegenerative disorders. Front Immunol 2023; 14:1233870. [PMID: 37575227 PMCID: PMC10416633 DOI: 10.3389/fimmu.2023.1233870] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
CD8+ lymphocytes are adaptive immunity cells with the particular function to directly kill the target cell following antigen recognition in the context of MHC class I. In addition, CD8+ T cells may release pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), and a plethora of other cytokines and chemoattractants modulating immune and inflammatory responses. A role for CD8+ T cells has been suggested in aging and several diseases of the central nervous system (CNS), including Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, limbic encephalitis-induced temporal lobe epilepsy and Susac syndrome. Here we discuss the phenotypic and functional alterations of CD8+ T cell compartment during these conditions, highlighting similarities and differences between CNS disorders. Particularly, we describe the pathological changes in CD8+ T cell memory phenotypes emphasizing the role of senescence and exhaustion in promoting neuroinflammation and neurodegeneration. We also discuss the relevance of trafficking molecules such as selectins, mucins and integrins controlling the extravasation of CD8+ T cells into the CNS and promoting disease development. Finally, we discuss how CD8+ T cells may induce CNS tissue damage leading to neurodegeneration and suggest that targeting detrimental CD8+ T cells functions may have therapeutic effect in CNS disorders.
Collapse
Affiliation(s)
- Eleonora Terrabuio
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | | | | |
Collapse
|
29
|
Walker KA, Le Page LM, Terrando N, Duggan MR, Heneka MT, Bettcher BM. The role of peripheral inflammatory insults in Alzheimer's disease: a review and research roadmap. Mol Neurodegener 2023; 18:37. [PMID: 37277738 PMCID: PMC10240487 DOI: 10.1186/s13024-023-00627-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 05/24/2023] [Indexed: 06/07/2023] Open
Abstract
Peripheral inflammation, defined as inflammation that occurs outside the central nervous system, is an age-related phenomenon that has been identified as a risk factor for Alzheimer's disease. While the role of chronic peripheral inflammation has been well characterized in the context of dementia and other age-related conditions, less is known about the neurologic contribution of acute inflammatory insults that take place outside the central nervous system. Herein, we define acute inflammatory insults as an immune challenge in the form of pathogen exposure (e.g., viral infection) or tissue damage (e.g., surgery) that causes a large, yet time-limited, inflammatory response. We provide an overview of the clinical and translational research that has examined the connection between acute inflammatory insults and Alzheimer's disease, focusing on three categories of peripheral inflammatory insults that have received considerable attention in recent years: acute infection, critical illness, and surgery. Additionally, we review immune and neurobiological mechanisms which facilitate the neural response to acute inflammation and discuss the potential role of the blood-brain barrier and other components of the neuro-immune axis in Alzheimer's disease. After highlighting the knowledge gaps in this area of research, we propose a roadmap to address methodological challenges, suboptimal study design, and paucity of transdisciplinary research efforts that have thus far limited our understanding of how pathogen- and damage-mediated inflammatory insults may contribute to Alzheimer's disease. Finally, we discuss how therapeutic approaches designed to promote the resolution of inflammation may be used following acute inflammatory insults to preserve brain health and limit progression of neurodegenerative pathology.
Collapse
Affiliation(s)
- Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute On Aging. Baltimore, Baltimore, MD, USA.
| | - Lydia M Le Page
- Departments of Physical Therapy and Rehabilitation Science, and Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Niccolò Terrando
- Department of Anesthesiology, Cell Biology and Immunology, Duke University Medical Center, Durham, NC, USA
| | - Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute On Aging. Baltimore, Baltimore, MD, USA
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Brianne M Bettcher
- Behavioral Neurology Section, Department of Neurology, University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
30
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley K, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531435. [PMID: 36945372 PMCID: PMC10028873 DOI: 10.1101/2023.03.07.531435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Major Histocompatibility Complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating Ribosome Affinity Purification-qPCR analysis of 3-6 and 18-22 month old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m , H2-D1 , H2-K1 , H2-M3 , H2-Q6 , and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I binding Leukocyte Immunoglobulin-like (Lilrs) and Paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell-autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A , suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M. Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Kevin Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Adeline H. Machalinski
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Hunter L. Porter
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Harris E. Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla Tooley
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Heather C. Rice
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Amanda L. Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J. Beckstead
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Ana J. Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Sarah R. Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Willard M. Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK USA
| |
Collapse
|
31
|
Fernando N, Gopalakrishnan J, Behensky A, Reich L, Liu C, Bass V, Bono M, Montgomery W, De Pace R, Mattapallil M, Nagarajan V, Brooks S, Maric D, Caspi RR, McGavern DB, Shih HY. Single-cell multiomic analysis reveals the involvement of Type I interferon-responsive CD8+ T cells in amyloid beta-associated memory loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.18.533293. [PMID: 37090642 PMCID: PMC10120715 DOI: 10.1101/2023.03.18.533293] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide, but there are limited therapeutic options and no current cure. While the involvement of microglia in AD has been highly appreciated, the role of other innate and adaptive immune cells remains largely unknown, partly due to their scarcity and heterogeneity. This study aimed to study non-microglial immune cells in wild type and AD-transgenic mouse brains across different ages. Our results uncovered the presence of a unique CD8+ T cell population that were selectively increased in aging AD mouse brains, here referred to as "disease-associated T cells (DATs)". These DATs were found to express an elevated tissue-resident memory and Type I interferon-responsive gene signature. Further analysis of aged AD mouse brains showed that these CD8+ T cells were not present in peripheral or meningeal tissues. Preventing CD8+ T cell development in AD-transgenic mice via genetic deletion of beta-2 microglobulin ( B2m ) led to a reduction of amyloid-β plaque formation in aged mice, and improved memory in AD-transgenic mice as early as four months of age. The integration of transcriptomic and epigenomic profiles at the single-cell level revealed potential transcription factors that reshape the regulomes of CD8+ T cells. These findings highlight a critical role for DATs in the progression of AD and provide a new avenue for treatment.
Collapse
|
32
|
Abstract
Microglia contribute to neurodegeneration through numerous mechanisms. In this issue of Neuron, Shi et al.1 identify a maladaptive innate-adaptive immune axis with CD8+ T cells, mediated through microglial CCL2/8 and CCR2/5, in radiation-induced brain injury and stroke. Their findings across species and injuries suggest broader implications for neurodegenerative conditions.
Collapse
|
33
|
Han PC, Hamlett ED. T Lymphocytes in the Pathogenesis and Progression of Alzheimer's Disease: Pursuing Direct Neuropathological Evidence. Curr Alzheimer Res 2023; 20:453-458. [PMID: 37670715 DOI: 10.2174/1567205020666230904151011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/09/2023] [Accepted: 07/04/2023] [Indexed: 09/07/2023]
Abstract
Multiple studies have proposed important roles of T cells in the pathogenesis of Alzheimer's disease. Given the successful application of immune-based therapy for cancer and a variety of diseases, T cell-modifying therapy becomes an attractive way to develop new therapies for Alzheimer's disease and perhaps neurodegenerative diseases in general. However, most of these studies address peripheral T cell responses, while direct pathological evidence documenting T cell infiltration relative to Alzheimer's disease pathological markers (i.e., amyloid plaque and neurofibrillary tangle) is sparse and at best, very preliminary in both human subjects and relevant animal models. Here, we concisely summarize the available pathological data that directly corresponds to T cell infiltration, critically analyze the current knowledge gaps, and thoughtfully propose several key recommendations for future research.
Collapse
Affiliation(s)
- Peng Cheng Han
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University, School of Medicine, Morgantown, WV 26501, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26501, USA
| | - Eric Daniel Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29401, USA
| |
Collapse
|
34
|
Lai Y, Lin P, Lin F, Chen M, Lin C, Lin X, Wu L, Zheng M, Chen J. Identification of immune microenvironment subtypes and signature genes for Alzheimer's disease diagnosis and risk prediction based on explainable machine learning. Front Immunol 2022; 13:1046410. [PMID: 36569892 PMCID: PMC9773397 DOI: 10.3389/fimmu.2022.1046410] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Background Using interpretable machine learning, we sought to define the immune microenvironment subtypes and distinctive genes in AD. Methods ssGSEA, LASSO regression, and WGCNA algorithms were used to evaluate immune state in AD patients. To predict the fate of AD and identify distinctive genes, six machine learning algorithms were developed. The output of machine learning models was interpreted using the SHAP and LIME algorithms. For external validation, four separate GEO databases were used. We estimated the subgroups of the immunological microenvironment using unsupervised clustering. Further research was done on the variations in immunological microenvironment, enhanced functions and pathways, and therapeutic medicines between these subtypes. Finally, the expression of characteristic genes was verified using the AlzData and pan-cancer databases and RT-PCR analysis. Results It was determined that AD is connected to changes in the immunological microenvironment. WGCNA revealed 31 potential immune genes, of which the greenyellow and blue modules were shown to be most associated with infiltrated immune cells. In the testing set, the XGBoost algorithm had the best performance with an AUC of 0.86 and a P-R value of 0.83. Following the screening of the testing set by machine learning algorithms and the verification of independent datasets, five genes (CXCR4, PPP3R1, HSP90AB1, CXCL10, and S100A12) that were closely associated with AD pathological biomarkers and allowed for the accurate prediction of AD progression were found to be immune microenvironment-related genes. The feature gene-based nomogram may provide clinical advantages to patients. Two immune microenvironment subgroups for AD patients were identified, subtype2 was linked to a metabolic phenotype, subtype1 belonged to the immune-active kind. MK-866 and arachidonyltrifluoromethane were identified as the top treatment agents for subtypes 1 and 2, respectively. These five distinguishing genes were found to be intimately linked to the development of the disease, according to the Alzdata database, pan-cancer research, and RT-PCR analysis. Conclusion The hub genes associated with the immune microenvironment that are most strongly associated with the progression of pathology in AD are CXCR4, PPP3R1, HSP90AB1, CXCL10, and S100A12. The hypothesized molecular subgroups might offer novel perceptions for individualized AD treatment.
Collapse
Affiliation(s)
- Yongxing Lai
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Peiqiang Lin
- Department of Neurology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Fan Lin
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chunjin Lin
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Xing Lin
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Lijuan Wu
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Mouwei Zheng
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China,*Correspondence: Jianhao Chen, ; Mouwei Zheng,
| | - Jianhao Chen
- Department of Rehabilitation Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,*Correspondence: Jianhao Chen, ; Mouwei Zheng,
| |
Collapse
|