1
|
Zhao W, Ma L, Deng D, Han L, Xu F, Zhang T, Wang Y, Huang S, Ding Y, Shu S, Chen X. BDNF-VGF Pathway Aggravates Incision Induced Acute Postoperative Pain via Upregulating the Neuroinflammation in Dorsal Root Ganglia. Mol Neurobiol 2025; 62:169-183. [PMID: 38829513 DOI: 10.1007/s12035-024-04249-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 05/13/2024] [Indexed: 06/05/2024]
Abstract
Approximately one-third of postoperative patients are troubled by postoperative pain. Effective treatments are still lacking. The aim of this study is to investigate the role of brain-derived neurotrophic factor (BDNF)-VGF (non-acronymic) in dorsal root ganglia (DRG) in postoperative pain. Pain behaviors were assessed through measurements of paw withdrawal threshold (PWT) and paw withdrawal latency (PWL). Transcriptome analysis was conducted to identify potential targets associated with postoperative pain. Western blotting, immunofluorescence, and ELISA were employed to further detect macrophage activation as well as the expression of BDNF, VGF, TNF-α, IL-1β, and IL-6. Results showed that plantar incision induced both mechanical and thermal hyperalgesia. Transcriptome analysis suggested that plantar incision caused upregulation of BDNF and VGF. The expressions of BDNF and VGF were upregulated in isolectin B4-positive (IB4+) and calcitonin gene-related peptide-positive (CGRP+) neurons, rather than neurofilament 200-positive (NF200+) neurons. The activation of BDNF-VGF pathway upregulated expression of IL-6, TNF-α, and IL-1β and promoted the activation of macrophages. In conclusion, BDNF-VGF pathway aggravates acute postoperative pain by promoting macrophage activation and pro-inflammatory cytokine production, which may provide a new target for the treatment of postoperative pain.
Collapse
Affiliation(s)
- Wenjing Zhao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Lulin Ma
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Feng Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Shaofang Shu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
2
|
Gedam M, Zheng H. Complement C3aR signaling: Immune and metabolic modulation and its impact on Alzheimer's disease. Eur J Immunol 2024; 54:e2350815. [PMID: 38778507 PMCID: PMC11305912 DOI: 10.1002/eji.202350815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia among the elderly population. Despite its widespread prevalence, our comprehension of the intricate mechanisms governing the pathogenesis of the disease remains incomplete, posing a challenge for the development of efficient therapies. Pathologically characterized by the presence of amyloid β plaques and neurofibrillary tau tangles, AD is also accompanied by the hyperactivation of glial cells and the immune system. The complement cascade, the evolutionarily conserved innate immune pathway, has emerged as a significant contributor to AD. This review focuses on one of the complement components, the C3a receptor (C3aR), covering its structure, ligand-receptor interaction, intracellular signaling and its functional consequences. Drawing insights from cellular and AD mouse model studies, we present the multifaceted role of complement C3aR signaling in AD and attempt to convey to the readers that C3aR acts as a crucial immune and metabolic modulator to influence AD pathogenesis. Building on this framework, the objective of this review is to inform future research endeavors and facilitate the development of therapeutic strategies for this challenging condition.
Collapse
Affiliation(s)
- Manasee Gedam
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Hui Zheng
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
3
|
Woo MS, Bal LC, Winschel I, Manca E, Walkenhorst M, Sevgili B, Sonner JK, Di Liberto G, Mayer C, Binkle-Ladisch L, Rothammer N, Unger L, Raich L, Hadjilaou A, Noli B, Manai AL, Vieira V, Meurs N, Wagner I, Pless O, Cocco C, Stephens SB, Glatzel M, Merkler D, Friese MA. The NR4A2/VGF pathway fuels inflammation-induced neurodegeneration via promoting neuronal glycolysis. J Clin Invest 2024; 134:e177692. [PMID: 39145444 PMCID: PMC11324305 DOI: 10.1172/jci177692] [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: 11/16/2023] [Accepted: 06/11/2024] [Indexed: 08/16/2024] Open
Abstract
A disturbed balance between excitation and inhibition (E/I balance) is increasingly recognized as a key driver of neurodegeneration in multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system. To understand how chronic hyperexcitability contributes to neuronal loss in MS, we transcriptionally profiled neurons from mice lacking inhibitory metabotropic glutamate signaling with shifted E/I balance and increased vulnerability to inflammation-induced neurodegeneration. This revealed a prominent induction of the nuclear receptor NR4A2 in neurons. Mechanistically, NR4A2 increased susceptibility to excitotoxicity by stimulating continuous VGF secretion leading to glycolysis-dependent neuronal cell death. Extending these findings to people with MS (pwMS), we observed increased VGF levels in serum and brain biopsies. Notably, neuron-specific deletion of Vgf in a mouse model of MS ameliorated neurodegeneration. These findings underscore the detrimental effect of a persistent metabolic shift driven by excitatory activity as a fundamental mechanism in inflammation-induced neurodegeneration.
Collapse
Affiliation(s)
- Marcel S. Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C. Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Winschel
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elias Manca
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Mark Walkenhorst
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bachar Sevgili
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K. Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Giovanni Di Liberto
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Unger
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandros Hadjilaou
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard-Nocht-Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - Barbara Noli
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Antonio L. Manai
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Hamburg, Germany
| | - Cristina Cocco
- Department of Biomedical Sciences, NEF-Laboratory, University of Cagliari, Monserrato, Cagliari, Italy
| | - Samuel B. Stephens
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Kuhrt LD, Motta E, Elmadany N, Weidling H, Fritsche-Guenther R, Efe IE, Cobb O, Chatterjee J, Boggs LG, Schnauß M, Diecke S, Semtner M, Anastasaki C, Gutmann DH, Kettenmann H. Neurofibromin 1 mutations impair the function of human induced pluripotent stem cell-derived microglia. Dis Model Mech 2023; 16:dmm049861. [PMID: 37990867 PMCID: PMC10740172 DOI: 10.1242/dmm.049861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant condition caused by germline mutations in the neurofibromin 1 (NF1) gene. Children with NF1 are prone to the development of multiple nervous system abnormalities, including autism and brain tumors, which could reflect the effect of NF1 mutation on microglia function. Using heterozygous Nf1-mutant mice, we previously demonstrated that impaired purinergic signaling underlies deficits in microglia process extension and phagocytosis in situ. To determine whether these abnormalities are also observed in human microglia in the setting of NF1, we leveraged an engineered isogenic series of human induced pluripotent stem cells to generate human microglia-like (hiMGL) cells heterozygous for three different NF1 gene mutations found in patients with NF1. Whereas all NF1-mutant and isogenic control hiMGL cells expressed classical microglia markers and exhibited similar transcriptomes and cytokine/chemokine release profiles, only NF1-mutant hiMGL cells had defects in P2X receptor activation, phagocytosis and motility. Taken together, these findings indicate that heterozygous NF1 mutations impair a subset of the functional properties of human microglia, which could contribute to the neurological abnormalities seen in children with NF1.
Collapse
Affiliation(s)
- Leonard D. Kuhrt
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Technology Platform Pluripotent Stem Cells, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Edyta Motta
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Nirmeen Elmadany
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), Clinical Cooperation Unit (CCU), Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim (MCTN), University of Heidelberg, 68167 Mannheim, Germany
| | - Hannah Weidling
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Raphaela Fritsche-Guenther
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, BIH Metabolomics Platform, 13353 Berlin, Germany
| | - Ibrahim E. Efe
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lucy G. Boggs
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marina Schnauß
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Sebastian Diecke
- Technology Platform Pluripotent Stem Cells, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Klinik für Augenheilkunde, Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China, 518000
| |
Collapse
|
5
|
Chen Z, Giotti B, Kaluzova M, Vallcorba MP, Rawat K, Price G, Herting CJ, Pinero G, Cristea S, Ross JL, Ackley J, Maximov V, Szulzewsky F, Thomason W, Marquez-Ropero M, Angione A, Nichols N, Tsankova NM, Michor F, Shayakhmetov DM, Gutmann DH, Tsankov AM, Hambardzumyan D. A paracrine circuit of IL-1β/IL-1R1 between myeloid and tumor cells drives genotype-dependent glioblastoma progression. J Clin Invest 2023; 133:e163802. [PMID: 37733448 PMCID: PMC10645395 DOI: 10.1172/jci163802] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/19/2023] [Indexed: 09/23/2023] Open
Abstract
Monocytes and monocyte-derived macrophages (MDMs) from blood circulation infiltrate glioblastoma (GBM) and promote growth. Here, we show that PDGFB-driven GBM cells induce the expression of the potent proinflammatory cytokine IL-1β in MDM, which engages IL-1R1 in tumor cells, activates the NF-κB pathway, and subsequently leads to induction of monocyte chemoattractant proteins (MCPs). Thus, a feedforward paracrine circuit of IL-1β/IL-1R1 between tumors and MDM creates an interdependence driving PDGFB-driven GBM progression. Genetic loss or locally antagonizing IL-1β/IL-1R1 leads to reduced MDM infiltration, diminished tumor growth, and reduced exhausted CD8+ T cells and thereby extends the survival of tumor-bearing mice. In contrast to IL-1β, IL-1α exhibits antitumor effects. Genetic deletion of Il1a/b is associated with decreased recruitment of lymphoid cells and loss-of-interferon signaling in various immune populations and subsets of malignant cells and is associated with decreased survival time of PDGFB-driven tumor-bearing mice. In contrast to PDGFB-driven GBM, Nf1-silenced tumors have a constitutively active NF-κB pathway, which drives the expression of MCPs to recruit monocytes into tumors. These results indicate local antagonism of IL-1β could be considered as an effective therapy specifically for proneural GBM.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Milota Kaluzova
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Department of Neurology, Rutgers University, New Brunswick, New Jersey, USA
| | - Montse Puigdelloses Vallcorba
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Kavita Rawat
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Gabrielle Price
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Cameron J. Herting
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Gonzalo Pinero
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - James L. Ross
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Emory University Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, Georgia, USA
| | - James Ackley
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Victor Maximov
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Frank Szulzewsky
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Wes Thomason
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Mar Marquez-Ropero
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Angelo Angione
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | | | - Nadejda M. Tsankova
- Department of Pathology and Molecular and Cell-Based Medicine, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- The Ludwig Center at Harvard, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Dmitry M. Shayakhmetov
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Lowance Center for Human Immunology and Emory Vaccine Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexander M. Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Neurosurgery and
| |
Collapse
|
6
|
MohanKumar SMJ, Murugan A, Palaniyappan A, MohanKumar PS. Role of cytokines and reactive oxygen species in brain aging. Mech Ageing Dev 2023; 214:111855. [PMID: 37541628 PMCID: PMC10528856 DOI: 10.1016/j.mad.2023.111855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Aging is a complex process that produces profound effects on the brain. Although a number of external factors can promote the initiation and progression of brain aging, peripheral and central changes in the immune cells with time, also play an important role. Immunosenescence, which is an age-associated decline in immune function and Inflammaging, a low-grade inflammatory state in the aging brain contribute to an elevation in cytokine and reactive oxygen species production. In this review, we focus on the pro-inflammatory state that is established in the brain as a consequence of these two phenomena and the resulting detrimental changes in brain structure, function and repair that lead to a decline in central and neuroendocrine function.
Collapse
Affiliation(s)
- Sheba M J MohanKumar
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.
| | - Abarna Murugan
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Arunkumar Palaniyappan
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Puliyur S MohanKumar
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
7
|
Guneykaya D, Ugursu B, Logiacco F, Popp O, Feiks MA, Meyer N, Wendt S, Semtner M, Cherif F, Gauthier C, Madore C, Yin Z, Çınar Ö, Arslan T, Gerevich Z, Mertins P, Butovsky O, Kettenmann H, Wolf SA. Sex-specific microglia state in the Neuroligin-4 knock-out mouse model of autism spectrum disorder. Brain Behav Immun 2023; 111:61-75. [PMID: 37001827 PMCID: PMC10330133 DOI: 10.1016/j.bbi.2023.03.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 03/15/2023] [Accepted: 03/27/2023] [Indexed: 04/10/2023] Open
Abstract
Neuroligin-4 (NLGN4) loss-of-function mutations are associated with monogenic heritable autism spectrum disorder (ASD) and cause alterations in both synaptic and behavioral phenotypes. Microglia, the resident CNS macrophages, are implicated in ASD development and progression. Here we studied the impact of NLGN4 loss in a mouse model, focusing on microglia phenotype and function in both male and female mice. NLGN4 depletion caused lower microglia density, less ramified morphology, reduced response to injury and purinergic signaling specifically in the hippocampal CA3 region predominantly in male mice. Proteomic analysis revealed disrupted energy metabolism in male microglia and provided further evidence for sexual dimorphism in the ASD associated microglial phenotype. In addition, we observed impaired gamma oscillations in a sex-dependent manner. Lastly, estradiol application in male NLGN4-/- mice restored the altered microglial phenotype and function. Together, these results indicate that loss of NLGN4 affects not only neuronal network activity, but also changes the microglia state in a sex-dependent manner that could be targeted by estradiol treatment.
Collapse
Affiliation(s)
- Dilansu Guneykaya
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Neurobiology, Harvard Medical School, Boston, USA
| | - Bilge Ugursu
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Francesca Logiacco
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Popp
- Proteomics Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany
| | - Maria Almut Feiks
- Institute of Neurophysiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Niklas Meyer
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Stefan Wendt
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Marcus Semtner
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Fatma Cherif
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christian Gauthier
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Charlotte Madore
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Zhuoran Yin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Özcan Çınar
- Molecular Immunotherapy, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany
| | - Taner Arslan
- Department of Oncology and Pathology, Karolinska Institutet, Science for Life Laboratory, Solna, Sweden
| | - Zoltan Gerevich
- Institute of Neurophysiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute of Health, Berlin, Germany
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Germany
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Susanne A Wolf
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| |
Collapse
|
8
|
Quinn J, Ethier EC, Novielli A, Malone A, Ramirez CE, Salloum L, Trombetta BA, Kivisäkk P, Bremang M, Selzer S, Fournier M, Das S, Xing Y, Arnold SE, Carlyle BC. Cerebrospinal Fluid and Brain Proteoforms of the Granin Neuropeptide Family in Alzheimer's Disease. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:649-667. [PMID: 36912488 PMCID: PMC10080684 DOI: 10.1021/jasms.2c00341] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/11/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The granin neuropeptide family is composed of acidic secretory signaling molecules that act throughout the nervous system to help modulate synaptic signaling and neural activity. Granin neuropeptides have been shown to be dysregulated in different forms of dementia, including Alzheimer's disease (AD). Recent studies have suggested that the granin neuropeptides and their protease-cleaved bioactive peptides (proteoforms) may act as both powerful drivers of gene expression and as a biomarker of synaptic health in AD. The complexity of granin proteoforms in human cerebrospinal fluid (CSF) and brain tissue has not been directly addressed. We developed a reliable nontryptic mass spectrometry assay to comprehensively map and quantify endogenous neuropeptide proteoforms in the brain and CSF of individuals diagnosed with mild cognitive impairment and dementia due to AD compared to healthy controls, individuals with preserved cognition despite AD pathology ("Resilient"), and those with impaired cognition but no AD or other discernible pathology ("Frail"). We drew associations between neuropeptide proteoforms, cognitive status, and AD pathology values. Decreased levels of VGF proteoforms were observed in CSF and brain tissue from individuals with AD compared to controls, while select proteoforms from chromogranin A showed the opposite effect. To address mechanisms of neuropeptide proteoform regulation, we showed that the proteases Calpain-1 and Cathepsin S can cleave chromogranin A, secretogranin-1, and VGF into proteoforms found in both the brain and CSF. We were unable to demonstrate differences in protease abundance in protein extracts from matched brains, suggesting that regulation may occur at the level of transcription.
Collapse
Affiliation(s)
- James
P. Quinn
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Elizabeth C. Ethier
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Angelo Novielli
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Aygul Malone
- Advanced
Proteomics Facility, Department of Biochemistry, University of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
| | - Christopher E. Ramirez
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Lauren Salloum
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Bianca A. Trombetta
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Pia Kivisäkk
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Michael Bremang
- Proteome
Sciences LLC, Frankfurt am Main, Hessen 60438, Germany
| | - Stefan Selzer
- Proteome
Sciences LLC, Frankfurt am Main, Hessen 60438, Germany
| | - Marjorie Fournier
- Advanced
Proteomics Facility, Department of Biochemistry, University of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
| | - Sudeshna Das
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Yaoyi Xing
- Department
of Physiology, Anatomy & Genetics, University
of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
- Kavli
Institute for Nanoscience Discovery, University
of Oxford, Oxford OX1 3QU, United
Kingdom
| | - Steven E. Arnold
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Becky C. Carlyle
- Massachusetts
General Hospital Department of Neurology, Harvard Medical School, Boston, Massachusetts 02129, United States
- Department
of Physiology, Anatomy & Genetics, University
of Oxford, Oxford, Oxfordshire OX1 3QU, United Kingdom
- Kavli
Institute for Nanoscience Discovery, University
of Oxford, Oxford OX1 3QU, United
Kingdom
| |
Collapse
|
9
|
Gillis HL, Kalinina A, Xue Y, Yan K, Turcotte-Cardin V, Todd MAM, Young KG, Lagace D, Picketts DJ. VGF is required for recovery after focal stroke. Exp Neurol 2023; 362:114326. [PMID: 36682400 DOI: 10.1016/j.expneurol.2023.114326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/06/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
The high incidence of ischemic stroke worldwide and poor efficacy of neuroprotective drugs has increased the need for novel therapies in stroke recovery. Transcription of the neurosecretory protein VGF (non-acronym) is enhanced following ischemic stroke and proposed to be important for stroke recovery. To determine the requirement for VGF in recovery, we created Vgffl/fl:Nestin-Cre conditional knockout (Vgf cKO) mice and induced a photothrombotic focal ischemic stroke. Naïve Vgf cKO mice had significant less body weight in the absence of gross defects in brain size, cortical lamination, or deficits in locomotor activity compared to wildtype controls. Following a focal stroke, the Vgf cKO mice had greater deficits including impaired recovery of forepaw motor deficits at 2- and 4-weeks post stroke. The increase in deficits occurred in the absence of any difference in lesion size and was accompanied by a striking loss of stroke-induced migration of SVZ-derived immature neurons to the peri-infarct region. Importantly, exogenous adenoviral delivery of VGF (AdVGF) significantly improved recovery in the Vgf cKO mice and was able to rescue the immature neuron migration defect observed. Taken together, our results define a requirement for VGF in post stroke recovery and identify VGF peptides as a potential future therapeutic.
Collapse
Affiliation(s)
- Hannah L Gillis
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Departments of Biochemistry, Microbiology and Immunology, K1H 8M5, Canada
| | - Alena Kalinina
- Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Yingben Xue
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Keqin Yan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Valérie Turcotte-Cardin
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Matthew A M Todd
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Departments of Biochemistry, Microbiology and Immunology, K1H 8M5, Canada
| | - Kevin G Young
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Diane Lagace
- Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - David J Picketts
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Departments of Biochemistry, Microbiology and Immunology, K1H 8M5, Canada; Departments of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
10
|
Logiacco F, Grzegorzek LC, Cordell EC, Popp O, Mertins P, Gutmann DH, Kettenmann H, Semtner M. Neurofibromatosis type 1-dependent alterations in mouse microglia function are not cell-intrinsic. Acta Neuropathol Commun 2023; 11:36. [PMID: 36890585 PMCID: PMC9996880 DOI: 10.1186/s40478-023-01525-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 02/05/2023] [Indexed: 03/10/2023] Open
Abstract
We previously discovered a sex-by-genotype defect in microglia function using a heterozygous germline knockout mouse model of Neurofibromatosis type 1 (Nf1 ± mice), in which only microglia from male Nf1 ± mice exhibited defects in purinergic signaling. Herein, we leveraged an unbiased proteomic approach to demonstrate that male, but not female, heterozygous Nf1 ± microglia exhibit differences in protein expression, which largely reflect pathways involved in cytoskeletal organization. In keeping with these predicted defects in cytoskeletal function, only male Nf1 ± microglia had reduced process arborization and surveillance capacity. To determine whether these microglial defects were cell autonomous or reflected adaptive responses to Nf1 heterozygosity in other cells in the brain, we generated conditional microglia Nf1-mutant knockout mice by intercrossing Nf1flox/flox with Cx3cr1-CreER mice (Nf1flox/wt; Cx3cr1-CreER mice, Nf1MG ± mice). Surprisingly, neither male nor female Nf1MG ± mouse microglia had impaired process arborization or surveillance capacity. In contrast, when Nf1 heterozygosity was generated in neurons, astrocytes and oligodendrocytes by intercrossing Nf1flox/flox with hGFAP-Cre mice (Nf1flox/wt; hGFAP-Cre mice, Nf1GFAP ± mice), the microglia defects found in Nf1 ± mice were recapitulated. Collectively, these data reveal that Nf1 ± sexually dimorphic microglia abnormalities are likely not cell-intrinsic properties, but rather reflect a response to Nf1 heterozygosity in other brain cells.
Collapse
Affiliation(s)
- Francesca Logiacco
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Laura Cathleen Grzegorzek
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Elizabeth C Cordell
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Oliver Popp
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany.
| |
Collapse
|
11
|
Hu X, Zhu L, Ouyang Q, Wang J, Hu J, Hu B, Hu S, He H, Li L, Liu H, Wang J. Comparative transcriptome analysis identified crucial genes and pathways affecting sperm motility in the reproductive tract of drakes with different libido. Poult Sci 2023; 102:102560. [PMID: 36881978 PMCID: PMC9993030 DOI: 10.1016/j.psj.2023.102560] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/21/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Libido can affect the semen quality of male, and the sperm motility in semen quality parameters is a reliable index to evaluate the fertility of male. In drakes, the sperm motility is gradually acquired in testis, epididymis, and spermaduct. However, the relationship between libido and sperm motility in drakes has not been reported and the mechanisms of testis, epididymis, and spermaduct regulating the sperm motility of drakes are unclear. Therefore, the purpose of the present study was to compare the semen quality of drakes with libido level 4 (LL4) and libido level 5 (LL5), and tried to identify the mechanisms regulating the sperm motility in drakes by performing RNA-seq in testis, epididymis, and spermaduct. Phenotypically, the sperm motility of drakes (P < 0.01), weight of testis (P < 0.05), and organ index of epididymis (P < 0.05) in the LL5 group were significantly better than those in LL4 group. Moreover, compared with the LL4 group, the ductal square of seminiferous tubule (ST) in testis was significantly bigger in the LL5 group (P < 0.05), and the seminiferous epithelial thickness (P < 0.01) of ST in testis and lumenal diameter (P < 0.05) of ductuli conjugentes/dutus epididymidis in epididymis were significantly longer in the LL5 group. In transcriptional regulation, in addition to KEGG pathways related to metabolism and oxidative phosphorylation, lots of KEGG pathways associated with immunity, proliferation, and signaling were also significantly enriched in testis, epididymis, and spermaduct, respectively. Furthermore, through the integrated analysis of coexpression network and protein-protein interaction network, 3 genes (including COL11A1, COL14A1, and C3AR1) involved in protein digestion and absorption pathway and Staphylococcus aureus infection pathway were identified in testis, 2 genes (including BUB1B and ESPL1) involved in cell cycle pathway were identified in epididymis, and 13 genes (including DNAH1, DNAH3, DNAH7, DNAH10, DNAH12, DNAI1, DNAI2, DNALI1, NTF3, ITGA1, TLR2, RELN, and PAK1) involved in Huntington disease pathway and PI3K-Akt signaling pathway were identified in spermaduct. These genes could play crucial roles in the sperm motility of drakes with different libido, and all data the present study obtained will provide new insights into the molecular mechanisms regulating sperm motility of drakes.
Collapse
Affiliation(s)
- Xinyue Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Lipeng Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Junqi Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130 Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Li XX, Lee JD, Lee HS, Clark RJ, Woodruff TM. TLQP-21 is a low potency partial C3aR activator on human primary macrophages. Front Immunol 2023; 14:1086673. [PMID: 36776827 PMCID: PMC9909341 DOI: 10.3389/fimmu.2023.1086673] [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: 11/01/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
TLQP-21 is a 21-amino acid neuropeptide derived from the VGF precursor protein. TLQP-21 is expressed in the nervous system and neuroendocrine glands, and demonstrates pleiotropic roles including regulating metabolism, nociception and microglial functions. Several possible receptors for TLQP-21 have been identified, with complement C3a receptor (C3aR) being the most commonly reported. However, few studies have characterised the activity of TLQP-21 in immune cells, which represent the major cell type expressing C3aR. In this study, we therefore aimed to define the activity of both human and mouse TLQP-21 on cell signalling in primary human and mouse macrophages. We first confirmed that TLQP-21 induced ERK signalling in CHO cells overexpressing human C3aR, and did not activate human C5aR1 or C5aR2. TLQP-21 mediated ERK signalling was also observed in primary human macrophages. However, the potency for human TLQP-21 was 135,000-fold lower relative to C3a, and only reached 45% at the highest dose tested (10 μM). Unlike in humans, mouse TLQP-21 potently triggered ERK signalling in murine macrophages, reaching near full activation, but at ~10-fold reduced potency compared to C3a. We further confirmed the C3aR dependency of the TLQP-21 activities. Our results reveal significant discrepancy in TLQP-21 C3aR activity between human and murine receptors, with mouse TLQP-21 being consistently more potent than the human counterpart in both systems. Considering the supraphysiological concentrations of hTLQP-21 needed to only partially activate macrophages, it is likely that the actions of TLQP-21, at least in these immune cells, may not be mediated by C3aR in humans.
Collapse
Affiliation(s)
- Xaria X Li
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Han S Lee
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Richard J Clark
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
13
|
Thalamocortical axons regulate neurogenesis and laminar fates in the early sensory cortex. Proc Natl Acad Sci U S A 2022; 119:e2201355119. [PMID: 35613048 PMCID: PMC9295754 DOI: 10.1073/pnas.2201355119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
This study addresses how the cerebral cortex is partitioned into specialized areas during development. Although both early embryonic patterning and postnatal synaptic input from sensory thalamic nuclei are known to be critical, early roles of thalamic axons in area-specific regulation of cortical neurogenesis are poorly understood. We examined this by developing a genetic mouse model in which thalamocortical projections fail to properly form during embryogenesis, and found these axons are required not only for an enhanced production of superficial layer neurons but also for promoting the layer 4 cell fate, a hallmark of the primary sensory cortex. These findings provide a mechanism by which thalamocortical axons complement the intrinsic programs of neurogenesis and early fate specification. Area-specific axonal projections from the mammalian thalamus shape unique cellular organization in target areas in the adult neocortex. How these axons control neurogenesis and early neuronal fate specification is poorly understood. By using mutant mice lacking the majority of thalamocortical axons, we show that these axons are required for the production and specification of the proper number of layer 4 neurons in primary sensory areas by the neonatal stage. Part of these area-specific roles is played by the thalamus-derived molecule, VGF. Our work reveals that extrinsic cues from sensory thalamic projections have an early role in the formation of cortical cytoarchitecture by enhancing the production and specification of layer 4 neurons.
Collapse
|
14
|
Jiang PP, Peng SS, Pankratova S, Luo P, Zhou P, Chen Y. Proteins Involved in Synaptic Plasticity Are Downregulated in the Cerebrospinal Fluid of Infants With Clinical Sepsis Complicated by Neuroinflammation. Front Cell Neurosci 2022; 16:887212. [PMID: 35634471 PMCID: PMC9130476 DOI: 10.3389/fncel.2022.887212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Newborn infants are prone to sepsis and related inflammation of different organs. Neuroinflammation has been associated with long-term adverse neuronal (neuropsychiatric/neurodegenerative) outcomes, including attention deficit hyperactivity disorder (ADHD) or even Alzheimer's disease. Despite a vast number of findings on sepsis-induced inflammatory responses in the central nervous system (CNS), how neuroinflammation affects brain development remains largely elusive. In this study, neonates with clinical sepsis and screened for meningitis were included and classified by the neuroinflammation status based on cerebrospinal fluid (CSF) parameters (INF vs. NOINF). CSF samples collected from clinical screening were subjected to proteomics analysis. Proteins with differential abundance were subjected to enrichment analysis to reveal affected biological pathways. INF and NOINF infants had similar demographic data and hematological and biochemical parameters in blood and CSF. The CSF proteomes were essentially different between the two groups. All 65 proteins with differential abundance showed lower abundance in the INF group and functionally covered pivotal developmental processes, including axonal and synaptic function and extracellular homeostasis. CSF proteins, PTPRZ1 and IGFBP4, were correlated with C-reactive protein (CRP) and ratios of immature/total neutrophils in blood. In general, a substantial change in the CSF protein profile was found under neuroinflammation, and these changes are related to systemic conditions. The results suggest that changes in CSF proteins may be involved in sepsis-affected neurodevelopment, such as disturbances in circuit formation, which has the potential to predispose neonates to long-term adverse outcomes.
Collapse
Affiliation(s)
- Ping-Ping Jiang
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shan-Shan Peng
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Stanislava Pankratova
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ping Luo
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ping Zhou
- Department of Neonatology, Bao'an Women and Children's Hospital, Jinan University, Shenzhen, China
| | - You Chen
- Department of Neonatology, Bao'an Women and Children's Hospital, Jinan University, Shenzhen, China
| |
Collapse
|
15
|
Sahu BS, Nguyen ME, Rodriguez P, Pallais JP, Ghosh V, Razzoli M, Sham YY, Salton SR, Bartolomucci A. The molecular identity of the TLQP-21 peptide receptor. Cell Mol Life Sci 2021; 78:7133-7144. [PMID: 34626205 PMCID: PMC8629782 DOI: 10.1007/s00018-021-03944-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022]
Abstract
The TLQP-21 neuropeptide has been implicated in functions as diverse as lipolysis, neurodegeneration and metabolism, thus suggesting an important role in several human diseases. Three binding targets have been proposed for TLQP-21: C3aR1, gC1qR and HSPA8. The aim of this review is to critically evaluate the molecular identity of the TLQP-21 receptor and the proposed multi-receptor mechanism of action. Several studies confirm a critical role for C3aR1 in TLQP-21 biological activity and a largely conserved mode of binding, receptor activation and signaling with C3a, its first-identified endogenous ligand. Conversely, data supporting a role of gC1qR and HSPA8 in TLQP-21 activity remain limited, with no signal transduction pathways being described. Overall, C3aR1 is the only receptor for which a necessary and sufficient role in TLQP-21 activity has been confirmed thus far. This conclusion calls into question the validity of a multi-receptor mechanism of action for TLQP-21 and should inform future studies.
Collapse
Affiliation(s)
- Bhavani S Sahu
- National Brain Research Centre, NH-8, Manesar, Gurugram, Haryana, 122052, India
| | - Megin E Nguyen
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, USA
| | - Pedro Rodriguez
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
| | - Jean Pierre Pallais
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
| | - Vinayak Ghosh
- National Brain Research Centre, NH-8, Manesar, Gurugram, Haryana, 122052, India
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
| | - Yuk Y Sham
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, USA
| | - Stephen R Salton
- Departments of Neuroscience and Geriatrics and Palliative Medicine, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
16
|
Quinn JP, Kandigian SE, Trombetta BA, Arnold SE, Carlyle BC. VGF as a biomarker and therapeutic target in neurodegenerative and psychiatric diseases. Brain Commun 2021; 3:fcab261. [PMID: 34778762 PMCID: PMC8578498 DOI: 10.1093/braincomms/fcab261] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Neurosecretory protein VGF (non-acronymic) belongs to the granin family of neuropeptides. VGF and VGF-derived peptides have been repeatedly identified in well-powered and well-designed multi-omic studies as dysregulated in neurodegenerative and psychiatric diseases. New therapeutics is urgently needed for these devastating and costly diseases, as are new biomarkers to improve disease diagnosis and mechanistic understanding. From a list of 537 genes involved in Alzheimer's disease pathogenesis, VGF was highlighted by the Accelerating Medicines Partnership in Alzheimer's disease as the potential therapeutic target of greatest interest. VGF levels are consistently decreased in brain tissue and CSF samples from patients with Alzheimer's disease compared to controls, and its levels correlate with disease severity and Alzheimer's disease pathology. In the brain, VGF exists as multiple functional VGF-derived peptides. Full-length human VGF1-615 undergoes proteolytic processing by prohormone convertases and other proteases in the regulated secretory pathway to produce at least 12 active VGF-derived peptides. In cell and animal models, these VGF-derived peptides have been linked to energy balance regulation, neurogenesis, synaptogenesis, learning and memory, and depression-related behaviours throughout development and adulthood. The C-terminal VGF-derived peptides, TLQP-62 (VGF554-615) and TLQP-21 (VGF554-574) have differential effects on Alzheimer's disease pathogenesis, neuronal and microglial activity, and learning and memory. TLQP-62 activates neuronal cell-surface receptors and regulates long-term hippocampal memory formation. TLQP-62 also prevents immune-mediated memory impairment, depression-like and anxiety-like behaviours in mice. TLQP-21 binds to microglial cell-surface receptors, triggering microglial chemotaxis and phagocytosis. These actions were reported to reduce amyloid-β plaques and decrease neuritic dystrophy in a transgenic mouse model of familial Alzheimer's disease. Expression differences of VGF-derived peptides have also been associated with frontotemporal lobar dementias, amyotrophic lateral sclerosis, Lewy body diseases, Huntington's disease, pain, schizophrenia, bipolar disorder, depression and antidepressant response. This review summarizes current knowledge and highlights questions for future investigation regarding the roles of VGF and its dysregulation in neurodegenerative and psychiatric disease. Finally, the potential of VGF and VGF-derived peptides as biomarkers and novel therapeutic targets for neurodegenerative and psychiatric diseases is highlighted.
Collapse
Affiliation(s)
- James P Quinn
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Savannah E Kandigian
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Bianca A Trombetta
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven E Arnold
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Becky C Carlyle
- Department of Neurology, Alzheimer's Clinical & Translational Research Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
The complement cascade in the regulation of neuroinflammation, nociceptive sensitization, and pain. J Biol Chem 2021; 297:101085. [PMID: 34411562 PMCID: PMC8446806 DOI: 10.1016/j.jbc.2021.101085] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 01/13/2023] Open
Abstract
The complement cascade is a key component of the innate immune system that is rapidly recruited through a cascade of enzymatic reactions to enable the recognition and clearance of pathogens and promote tissue repair. Despite its well-understood role in immunology, recent studies have highlighted new and unexpected roles of the complement cascade in neuroimmune interaction and in the regulation of neuronal processes during development, aging, and in disease states. Complement signaling is particularly important in directing neuronal responses to tissue injury, neurotrauma, and nerve lesions. Under physiological conditions, complement-dependent changes in neuronal excitability, synaptic strength, and neurite remodeling promote nerve regeneration, tissue repair, and healing. However, in a variety of pathologies, dysregulation of the complement cascade leads to chronic inflammation, persistent pain, and neural dysfunction. This review describes recent advances in our understanding of the multifaceted cross-communication that takes place between the complement system and neurons. In particular, we focus on the molecular and cellular mechanisms through which complement signaling regulates neuronal excitability and synaptic plasticity in the nociceptive pathways involved in pain processing in both health and disease. Finally, we discuss the future of this rapidly growing field and what we believe to be the significant knowledge gaps that need to be addressed.
Collapse
|
18
|
De Schepper S, Crowley G, Hong S. Understanding microglial diversity and implications for neuronal function in health and disease. Dev Neurobiol 2021; 81:507-523. [PMID: 32757416 PMCID: PMC8438703 DOI: 10.1002/dneu.22777] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/21/2020] [Accepted: 07/31/2020] [Indexed: 12/22/2022]
Abstract
Genetic data implicate microglia as central players in brain health and disease, urging the need to better understand what microglia do in the brain. Microglia are critical partners in neuronal wiring and function during development and disease. Emerging literature suggests that microglia have diverse functional roles, raising the intriguing question of which functions of microglia become impaired in disease to undermine proper neuronal function. It is also becoming increasingly clear that microglia exist in heterogeneous cell states. Microglial cell states appear context-dependent, that is, age, sex, location, and health of their microenvironment; these are further influenced by external signaling factors including gut microbiota and lipid metabolites. These data altogether suggest that microglia exist in functional clusters that impact, and are impacted by, surrounding neuronal microenvironment. However, we still lack understanding of how we can translate microglia cell states into function. Here, we summarize the state-of-the-art on the diverse functions of microglia in relation to neuronal health. Then, we discuss heterogeneity during developing, healthy adult and diseased brains, and whether this may be predetermined by origin and/or regulated by local milieu. Finally, we propose that it is critical to gain high-resolution functional discernment into microglia-neuron interactions while preserving the spatial architecture of the tissue. Such insight will reveal specific targets for biomarker and therapeutic development toward microglia-neuron crosstalk in disease.
Collapse
Affiliation(s)
| | - Gerard Crowley
- UK Dementia Research InstituteUniversity College LondonLondonUK
| | - Soyon Hong
- UK Dementia Research InstituteUniversity College LondonLondonUK
| |
Collapse
|
19
|
Xia P, Logiacco F, Huang Y, Kettenmann H, Semtner M. Histamine triggers microglial responses indirectly via astrocytes and purinergic signaling. Glia 2021; 69:2291-2304. [PMID: 34080730 DOI: 10.1002/glia.24039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
Histamine is a monoaminergic neurotransmitter which is released within the entire brain from ascending axons originating in the tuberomammillary nucleus in a sleep state-dependent fashion. Besides the modulation of neuronal firing patterns, brain histamine levels are also thought to modulate functions of glial cells. Microglia are the innate immune cells and professional phagocytes of the central nervous system, and histamine was previously shown to have multiple effects on microglial functions in health and disease. Isolated microglia respond only to agonists of the Hrh2 subtype of histamine receptors (Hrh), and the expression of that isoform is confirmed by a metadata analysis of microglia transcriptomes. When we studied the effect of the histamine receptor isoforms in cortical and thalamic microglia by in situ live cell Ca2+ imaging using a novel, microglia-specific indicator mouse line, microglial cells respond to external histamine application mainly in a Hrh1-, and to a lower extent also in a Hrh2-dependent manner. The Hrh1 response was sensitive to blockers of purinergic P2ry12 receptors, and since Hrh1 expression was predominantly found in astrocytes, we suggest that the Hrh1 response in microglia is mediated by astrocyte ATP release and activation of P2ry12 receptors in microglia. Histamine also stimulates microglial phagocytic activity via Hrh1- and P2ry12-mediated signaling. Taken together, we provide evidence that histamine acts indirectly on microglial Ca2+ levels and phagocytic activity via astrocyte histamine receptor-controlled purinergic signaling.
Collapse
Affiliation(s)
- Pengfei Xia
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité-Universitätsmedizin, Berlin, Germany
| | - Francesca Logiacco
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Yimin Huang
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité-Universitätsmedizin, Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
20
|
Elmadany N, Logiacco F, Buonfiglioli A, Haage VC, Wright-Jin EC, Schattenberg A, Papawassiliou RM, Kettenmann H, Semtner M, Gutmann DH. Neurofibromatosis 1 - Mutant microglia exhibit sexually-dimorphic cyclic AMP-dependent purinergic defects. Neurobiol Dis 2020; 144:105030. [PMID: 32736084 DOI: 10.1016/j.nbd.2020.105030] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/09/2020] [Accepted: 07/23/2020] [Indexed: 12/30/2022] Open
Abstract
As critical regulators of brain homeostasis, microglia are influenced by numerous factors, including sex and genetic mutations. To study the impact of these factors on microglia biology, we employed genetically engineered mice that model Neurofibromatosis type 1 (NF1), a disorder characterized by clinically relevant sexually dimorphic differences. While microglia phagocytic activity was reduced in both male and female heterozygous Nf1 mutant (Nf1+/-) mice, purinergic control of phagocytosis was only affected in male Nf1+/- mice. ATP-induced P2Y-mediated membrane currents and P2RY12-dependent laser lesion-induced accumulation of microglial processes were also only impaired in male, but not female Nf1+/-, microglia. These defects resulted from Nf1+/- male-specific defects in cyclic AMP regulation, rather than from changes in purinergic receptor expression. Cyclic AMP elevation by phosphodiesterase blockade restored the male Nf1+/- microglia defects in P2Y-dependent membrane currents and process motility. Taken together, these data establish a sex-by-genotype interaction important to microglia function in the adult mouse brain.
Collapse
Affiliation(s)
- Nirmeen Elmadany
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 12169 Berlin, Germany
| | - Francesca Logiacco
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 12169 Berlin, Germany
| | - Alice Buonfiglioli
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Verena C Haage
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Elizabeth C Wright-Jin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander Schattenberg
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Roxane M Papawassiliou
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| | - David H Gutmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
21
|
Beckmann ND, Lin WJ, Wang M, Cohain AT, Charney AW, Wang P, Ma W, Wang YC, Jiang C, Audrain M, Comella PH, Fakira AK, Hariharan SP, Belbin GM, Girdhar K, Levey AI, Seyfried NT, Dammer EB, Duong D, Lah JJ, Haure-Mirande JV, Shackleton B, Fanutza T, Blitzer R, Kenny E, Zhu J, Haroutunian V, Katsel P, Gandy S, Tu Z, Ehrlich ME, Zhang B, Salton SR, Schadt EE. Multiscale causal networks identify VGF as a key regulator of Alzheimer's disease. Nat Commun 2020; 11:3942. [PMID: 32770063 PMCID: PMC7414858 DOI: 10.1038/s41467-020-17405-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 06/15/2020] [Indexed: 12/31/2022] Open
Abstract
Though discovered over 100 years ago, the molecular foundation of sporadic Alzheimer's disease (AD) remains elusive. To better characterize the complex nature of AD, we constructed multiscale causal networks on a large human AD multi-omics dataset, integrating clinical features of AD, DNA variation, and gene- and protein-expression. These probabilistic causal models enabled detection, prioritization and replication of high-confidence master regulators of AD-associated networks, including the top predicted regulator, VGF. Overexpression of neuropeptide precursor VGF in 5xFAD mice partially rescued beta-amyloid-mediated memory impairment and neuropathology. Molecular validation of network predictions downstream of VGF was also achieved in this AD model, with significant enrichment for homologous genes identified as differentially expressed in 5xFAD brains overexpressing VGF. Our findings support a causal role for VGF in protecting against AD pathogenesis and progression.
Collapse
Affiliation(s)
- Noam D Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wei-Jye Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ariella T Cohain
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander W Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Center for Statistical Genetics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ying-Chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Cheng Jiang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Mickael Audrain
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Phillip H Comella
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda K Fakira
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Siddharth P Hariharan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Gillian M Belbin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Girdhar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Allan I Levey
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T Seyfried
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc Duong
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - James J Lah
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jean-Vianney Haure-Mirande
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ben Shackleton
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Tomas Fanutza
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Eimear Kenny
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Vahram Haroutunian
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Psychiatry, JJ Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Pavel Katsel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Psychiatry, JJ Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, JJ Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Zhidong Tu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Stephen R Salton
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Sema4, Stamford, CT, 06902, USA.
| |
Collapse
|