1
|
Sell GL, Barrow SL, McAllister AK. Glutamate signaling and neuroligin/neurexin adhesion play opposing roles that are mediated by major histocompatibility complex I molecules in cortical synapse formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583626. [PMID: 38496590 PMCID: PMC10942384 DOI: 10.1101/2024.03.05.583626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Although neurons release neurotransmitter before contact, the role for this release in synapse formation remains unclear. Cortical synapses do not require synaptic vesicle release for formation 1-4 , yet glutamate clearly regulates glutamate receptor trafficking 5,6 and induces spine formation 7-11 . Using a culture system to dissect molecular mechanisms, we found that glutamate rapidly decreases synapse density specifically in young cortical neurons in a local and calcium-dependent manner through decreasing NMDAR transport and surface expression as well as co-transport with neuroligin (NL1). Adhesion between NL1 and neurexin 1 protects against this glutamate-induced synapse loss. Major histocompatibility I (MHCI) molecules are required for the effects of glutamate in causing synapse loss through negatively regulating NL1 levels. Thus, like acetylcholine at the NMJ, glutamate acts as a dispersal signal for NMDARs and causes rapid synapse loss unless opposed by NL1-mediated trans-synaptic adhesion. Together, glutamate, MHCI and NL1 mediate a novel form of homeostatic plasticity in young neurons that induces rapid changes in NMDARs to regulate when and where nascent glutamatergic synapses are formed.
Collapse
|
2
|
Djurišić M. Immune receptors and aging brain. Biosci Rep 2024; 44:BSR20222267. [PMID: 38299364 PMCID: PMC10866841 DOI: 10.1042/bsr20222267] [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: 07/20/2023] [Revised: 01/08/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024] Open
Abstract
Aging brings about a myriad of degenerative processes throughout the body. A decrease in cognitive abilities is one of the hallmark phenotypes of aging, underpinned by neuroinflammation and neurodegeneration occurring in the brain. This review focuses on the role of different immune receptors expressed in cells of the central and peripheral nervous systems. We will discuss how immune receptors in the brain act as sentinels and effectors of the age-dependent shift in ligand composition. Within this 'old-age-ligand soup,' some immune receptors contribute directly to excessive synaptic weakening from within the neuronal compartment, while others amplify the damaging inflammatory environment in the brain. Ultimately, chronic inflammation sets up a positive feedback loop that increases the impact of immune ligand-receptor interactions in the brain, leading to permanent synaptic and neuronal loss.
Collapse
Affiliation(s)
- Maja Djurišić
- Departments of Biology, Neurobiology, and Bio-X, Stanford University, Stanford, CA 94305, U.S.A
| |
Collapse
|
3
|
La Cognata V, D’Amico AG, Maugeri G, Morello G, Guarnaccia M, Magrì B, Aronica E, Alkon DL, D’Agata V, Cavallaro S. The ε-Isozyme of Protein Kinase C (PKCε) Is Impaired in ALS Motor Cortex and Its Pulse Activation by Bryostatin-1 Produces Long Term Survival in Degenerating SOD1-G93A Motor Neuron-like Cells. Int J Mol Sci 2023; 24:12825. [PMID: 37629005 PMCID: PMC10454105 DOI: 10.3390/ijms241612825] [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: 07/11/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and ultimately fatal neurodegenerative disease, characterized by a progressive depletion of upper and lower motor neurons (MNs) in the brain and spinal cord. The aberrant regulation of several PKC-mediated signal transduction pathways in ALS has been characterized so far, describing either impaired expression or altered activity of single PKC isozymes (α, β, ζ and δ). Here, we detailed the distribution and cellular localization of the ε-isozyme of protein kinase C (PKCε) in human postmortem motor cortex specimens and reported a significant decrease in both PKCε mRNA (PRKCE) and protein immunoreactivity in a subset of sporadic ALS patients. We furthermore investigated the steady-state levels of both pan and phosphorylated PKCε in doxycycline-activated NSC-34 cell lines carrying the human wild-type (WT) or mutant G93A SOD1 and the biological long-term effect of its transient agonism by Bryostatin-1. The G93A-SOD1 cells showed a significant reduction of the phosphoPKCε/panPKCε ratio compared to the WT. Moreover, a brief pulse activation of PKCε by Bryostatin-1 produced long-term survival in activated G93A-SOD1 degenerating cells in two different cell death paradigms (serum starvation and chemokines-induced toxicity). Altogether, the data support the implication of PKCε in ALS pathophysiology and suggests its pharmacological modulation as a potential neuroprotective strategy, at least in a subgroup of sporadic ALS patients.
Collapse
Affiliation(s)
- Valentina La Cognata
- Institute for Biomedical Research and Innovation, National Research Council, 95126 Catania, Italy
| | - Agata Grazia D’Amico
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Grazia Maugeri
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giovanna Morello
- Institute for Biomedical Research and Innovation, National Research Council, 95126 Catania, Italy
| | - Maria Guarnaccia
- Institute for Biomedical Research and Innovation, National Research Council, 95126 Catania, Italy
| | - Benedetta Magrì
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105 Amsterdam, The Netherlands
| | | | - Velia D’Agata
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council, 95126 Catania, Italy
| |
Collapse
|
4
|
Eyford BA, Lazarczyk MJ, Choi KB, Varghese M, Arora H, Kari S, Munro L, Pfeifer CG, Sowa A, Dickstein DR, Dickstein DL, Jefferies WA. Outside-in signaling through the major histocompatibility complex class-I cytoplasmic tail modulates glutamate receptor expression in neurons. Sci Rep 2023; 13:13079. [PMID: 37567897 PMCID: PMC10421907 DOI: 10.1038/s41598-023-38663-z] [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: 03/16/2023] [Accepted: 07/12/2023] [Indexed: 08/13/2023] Open
Abstract
The interplay between AMPA-type glutamate receptors (AMPARs) and major histocompatibility complex class I (MHC-I) proteins in regulating synaptic signaling is a crucial aspect of central nervous system (CNS) function. In this study, we investigate the significance of the cytoplasmic tail of MHC-I in synaptic signaling within the CNS and its impact on the modulation of synaptic glutamate receptor expression. Specifically, we focus on the Y321 to F substitution (Y321F) within the conserved cytoplasmic tyrosine YXXΦ motif, known for its dual role in endocytosis and cellular signaling of MHC-I. Our findings reveal that the Y321F substitution influences the expression of AMPAR subunits GluA2/3 and leads to alterations in the phosphorylation of key kinases, including Fyn, Lyn, p38, ERK1/2, JNK1/2/3, and p70 S6 kinase. These data illuminate the crucial role of MHC-I in AMPAR function and present a novel mechanism by which MHC-I integrates extracellular cues to modulate synaptic plasticity in neurons, which ultimately underpins learning and memory.
Collapse
Affiliation(s)
- Brett A Eyford
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Maciej J Lazarczyk
- Division of Institutional Measures, Department of Medical Direction and Quality, University Hospitals of Geneva, Geneva, Switzerland
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Kyung Bok Choi
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Merina Varghese
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Hitesh Arora
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Suresh Kari
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lonna Munro
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Cheryl G Pfeifer
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Allison Sowa
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Daniel R Dickstein
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Dara L Dickstein
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA.
- Department of Pathology, Uniformed Services University of Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), 6720A Rockledge Drive, Bethesda, MD, 20817, USA.
| | - Wilfred A Jefferies
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada.
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada.
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
5
|
Zhang Y, Yan AW, Boelen L, Hadcocks L, Salam A, Gispert DP, Spanos L, Bitria LM, Nemat-Gorgani N, Traherne JA, Roberts C, Koftori D, Taylor GP, Forton D, Norman PJ, Marsh SG, Busch R, Macallan DC, Asquith B. KIR-HLA interactions extend human CD8+ T cell lifespan in vivo. J Clin Invest 2023; 133:e169496. [PMID: 37071474 PMCID: PMC10266773 DOI: 10.1172/jci169496] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/05/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUNDThere is increasing evidence, in transgenic mice and in vitro, that inhibitory killer cell immunoglobulin-like receptors (iKIRs) can modulate T cell responses. Furthermore, we have previously shown that iKIRs are an important determinant of T cell-mediated control of chronic viral infection and that these results are consistent with an increase in the CD8+ T cell lifespan due to iKIR-ligand interactions. Here, we tested this prediction and investigated whether iKIRs affect T cell lifespan in humans in vivo.METHODSWe used stable isotope labeling with deuterated water to quantify memory CD8+ T cell survival in healthy individuals and patients with chronic viral infections.RESULTSWe showed that an individual's iKIR-ligand genotype was a significant determinant of CD8+ T cell lifespan: in individuals with 2 iKIR-ligand gene pairs, memory CD8+ T cells survived, on average, for 125 days; in individuals with 4 iKIR-ligand gene pairs, the memory CD8+ T cell lifespan doubled to 250 days. Additionally, we showed that this survival advantage was independent of iKIR expression by the T cell of interest and, further, that the iKIR-ligand genotype altered the CD8+ and CD4+ T cell immune aging phenotype.CONCLUSIONSTogether, these data reveal an unexpectedly large effect of iKIR genotype on T cell survival.FUNDINGWellcome Trust; Medical Research Council; EU Horizon 2020; EU FP7; Leukemia and Lymphoma Research; National Institute of Health Research (NIHR) Imperial Biomedical Research Centre; Imperial College Research Fellowship; National Institutes of Health; Jefferiss Trust.
Collapse
Affiliation(s)
- Yan Zhang
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | - Ada W.C. Yan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Lies Boelen
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Linda Hadcocks
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | - Arafa Salam
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | | | - Loiza Spanos
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
- School of Life and Health Sciences, University of Roehampton, London, United Kingdom
| | - Laura Mora Bitria
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Neda Nemat-Gorgani
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - James A. Traherne
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Chrissy Roberts
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Danai Koftori
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Graham P. Taylor
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- National Centre for Human Retrovirology, St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Daniel Forton
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
- Department of Gastroenterology and Hepatology, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Paul J. Norman
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Department of Biomedical Informatics and Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Steven G.E. Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
- UCL Cancer Institute, UCL, London, United Kingdom
| | - Robert Busch
- School of Life and Health Sciences, University of Roehampton, London, United Kingdom
| | - Derek C. Macallan
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Mora-Bitria L, Asquith B. Innate receptors modulating adaptive T cell responses: KIR-HLA interactions and T cell-mediated control of chronic viral infections. Immunogenetics 2023; 75:269-282. [PMID: 36719466 PMCID: PMC9887252 DOI: 10.1007/s00251-023-01293-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/02/2023] [Indexed: 02/01/2023]
Abstract
Killer-cell immunoglobulin-like receptors (KIRs) are mainly expressed on natural killer (NK) cells and are key regulators of innate immune responses. NK cells are the first responders in the face of infection and help promote placentation during pregnancy; the importance of KIRs in these NK-mediated processes is well-established. However, mounting evidence suggests that KIRs also have a prominent and long-lasting effect on the adaptive immune system. Here, we review the evidence for the impact of KIRs on T cell responses with a focus on the clinical significance of this interaction.
Collapse
Affiliation(s)
- Laura Mora-Bitria
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Becca Asquith
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
7
|
Cukier HN, Kim H, Griswold AJ, Codreanu SG, Prince LM, Sherrod SD, McLean JA, Dykxhoorn DM, Ess KC, Hedera P, Bowman AB, Neely MD. Genomic, transcriptomic, and metabolomic profiles of hiPSC-derived dopamine neurons from clinically discordant brothers with identical PRKN deletions. NPJ Parkinsons Dis 2022; 8:84. [PMID: 35768426 PMCID: PMC9243035 DOI: 10.1038/s41531-022-00346-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
We previously reported on two brothers who carry identical compound heterozygous PRKN mutations yet present with significantly different Parkinson's Disease (PD) clinical phenotypes. Juvenile cases demonstrate that PD is not necessarily an aging-associated disease. Indeed, evidence for a developmental component to PD pathogenesis is accumulating. Thus, we hypothesized that the presence of additional genetic modifiers, including genetic loci relevant to mesencephalic dopamine neuron development, could potentially contribute to the different clinical manifestations of the two brothers. We differentiated human-induced pluripotent stem cells (hiPSCs) derived from the two brothers into mesencephalic neural precursor cells and early postmitotic dopaminergic neurons and performed wholeexome sequencing and transcriptomic and metabolomic analyses. No significant differences in the expression of canonical dopamine neuron differentiation markers were observed. Yet our transcriptomic analysis revealed a significant downregulation of the expression of three neurodevelopmentally relevant cell adhesion molecules, CNTN6, CNTN4 and CHL1, in the cultures of the more severely affected brother. In addition, several HLA genes, known to play a role in neurodevelopment, were differentially regulated. The expression of EN2, a transcription factor crucial for mesencephalic dopamine neuron development, was also differentially regulated. We further identified differences in cellular processes relevant to dopamine metabolism. Lastly, wholeexome sequencing, transcriptomics and metabolomics data all revealed differences in glutathione (GSH) homeostasis, the dysregulation of which has been previously associated with PD. In summary, we identified genetic differences which could potentially, at least partially, contribute to the discordant clinical PD presentation of the two brothers.
Collapse
Affiliation(s)
- Holly N Cukier
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hyunjin Kim
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simona G Codreanu
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kevin C Ess
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter Hedera
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, University of Louisville, Louisville, KY, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA.
| | - M Diana Neely
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
8
|
Synergized regulation of NK cell education by NKG2A and specific Ly49 family members. Nat Commun 2019; 10:5010. [PMID: 31676749 PMCID: PMC6825122 DOI: 10.1038/s41467-019-13032-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 10/17/2019] [Indexed: 12/15/2022] Open
Abstract
Mice lacking MHC class-I (MHC-I) display severe defects in natural killer (NK) cell functional maturation, a process designated as “education”. Whether self-MHC-I specific Ly49 family receptors and NKG2A, which are closely linked within the NK gene complex (NKC) locus, are essential for NK cell education is still unclear. Here we show, using CRISPR/Cas9-mediated gene deletion, that mice lacking all members of the Ly49 family exhibit a moderate defect in NK cell activity, while mice lacking only two inhibitory Ly49 members, Ly49C and Ly49I, have comparable phenotypes. Furthermore, the deficiency of NKG2A, which recognizes non-classical MHC-Ib molecules, mildly impairs NK cell function. Notably, the combined deletion of NKG2A and the Ly49 family severely compromises the ability of NK cells to mediate “missing-self” and “induced-self” recognition. Therefore, our data provide genetic evidence supporting that NKG2A and the inhibitory members of Ly49 family receptors synergize to regulate NK cell education. MHC-I-induced signalling of various natural killer (NK) inhibitory receptors is critical for regulation NK cell education, but clear genetic evidence is still lacking. Here the authors generate multiple lines of mice differentially deficient in Ly49 family and/or NKG2A NK receptors, and find that self-MHCI specific Ly49 members and NKG2A synergize to regulate NK education.
Collapse
|
9
|
Morimoto K, Nakajima K. Role of the Immune System in the Development of the Central Nervous System. Front Neurosci 2019; 13:916. [PMID: 31551681 PMCID: PMC6735264 DOI: 10.3389/fnins.2019.00916] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/16/2019] [Indexed: 01/01/2023] Open
Abstract
The central nervous system (CNS) and the immune system are both intricate and highly organized systems that regulate the entire body, with both sharing certain common features in developmental mechanisms and operational modes. It is known that innate immunity-related molecules, such as cytokines, toll-like receptors, the complement family, and acquired immunity-related molecules, such as the major histocompatibility complex and antibody receptors, are also expressed in the brain and play important roles in brain development. Moreover, although the brain has previously been regarded as an immune-privileged site, it is known to contain lymphatic vessels. Not only microglia but also lymphocytes regulate cognition and play a vital role in the formation of neuronal circuits. This review provides an overview of the function of immune cells and immune molecules in the CNS, with particular emphasis on their effect on neural developmental processes.
Collapse
Affiliation(s)
- Keiko Morimoto
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Pronovost GN, Hsiao EY. Perinatal Interactions between the Microbiome, Immunity, and Neurodevelopment. Immunity 2019; 50:18-36. [PMID: 30650376 PMCID: PMC6447295 DOI: 10.1016/j.immuni.2018.11.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/17/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
The microbiome modulates host immune function across the gastrointestinal tract, peripheral lymphoid organs, and central nervous system. In this review, we highlight emerging evidence that microbial effects on select immune phenotypes arise developmentally, where the maternal and neonatal microbiome influence immune cell ontogeny in the offspring during gestation and early postnatal life. We further discuss roles for the perinatal microbiome and early-life immunity in regulating normal neurodevelopmental processes. In addition, we examine evidence that abnormalities in microbiota-neuroimmune interactions during early life are associated with altered risk of neurological disorders in humans. Finally, we conclude by evaluating the potential implications of microbiota-immune interventions for neurological conditions. Continued progress toward dissecting mechanistic interactions between the perinatal microbiota, immune system, and nervous system might uncover fundamental insights into how developmental interactions across physiological systems inform later-life health and disease.
Collapse
Affiliation(s)
- Geoffrey N Pronovost
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Abstract
Epidemiological studies and mouse models suggest that maternal immune activation, induced clinically through prenatal exposure to one of several infectious diseases, is a risk factor in the development of schizophrenia. This is supported by the strong genetic association established by genome wide association studies (GWAS) between the human leukocyte antigen (HLA) locus and schizophrenia. HLA proteins (also known in mice as the major histocompatibility complex; MHC) are mediators of the T-lymphocyte responses, and genetic variability is well-established as a risk factor for autoimmune diseases and susceptibility to infectious diseases. Taken together, the findings strongly suggest that schizophrenia risk in a subgroup of patients is caused by an infectious disease, and/or an autoimmune phenomenon. However, this view may be overly simplistic. First, MHC proteins have a non-immune effect on synaptogenesis by modulating synaptic pruning by microglia and other mechanisms, suggesting that genetic variability could be compromising this physiological process. Second, some GWAS signals in the HLA locus map near non-HLA genes, such as the histone gene cluster. On the other hand, recent GWAS data show association signals near B-lymphocyte enhancers, which lend support for an infectious disease etiology. Thus, although the genetic findings implicating the HLA locus are very robust, how genetic variability in this region leads to schizophrenia remains to be elucidated.
Collapse
Affiliation(s)
- Ryan Mokhtari
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA
| | - Herbert M Lachman
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA; Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA; Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA; Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York, USA
| |
Collapse
|
12
|
Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
13
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
14
|
Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
15
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
16
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
17
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
18
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
19
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016; 10:105. [PMID: 27846841 PMCID: PMC5111260 DOI: 10.1186/s12918-016-0366-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 11/06/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND Individuals with 22q11.2 Deletion Syndrome (22q11.2 DS) are a specific high-risk group for developing schizophrenia (SZ), schizoaffective disorder (SAD) and autism spectrum disorders (ASD). Several genes in the deleted region have been implicated in the development of SZ, e.g., PRODH and DGCR8. However, the mechanistic connection between these genes and the neuropsychiatric phenotype remains unclear. To elucidate the molecular consequences of 22q11.2 deletion in early neural development, we carried out RNA-seq analysis to investigate gene expression in early differentiating human neurons derived from induced pluripotent stem cells (iPSCs) of 22q11.2 DS SZ and SAD patients. METHODS Eight cases (ten iPSC-neuron samples in total including duplicate clones) and seven controls (nine in total including duplicate clones) were subjected to RNA sequencing. Using a systems level analysis, differentially expressed genes/gene-modules and pathway of interests were identified. Lastly, we related our findings from in vitro neuronal cultures to brain development by mapping differentially expressed genes to BrainSpan transcriptomes. RESULTS We observed ~2-fold reduction in expression of almost all genes in the 22q11.2 region in SZ (37 genes reached p-value < 0.05, 36 of which reached a false discovery rate < 0.05). Outside of the deleted region, 745 genes showed significant differences in expression between SZ and control neurons (p < 0.05). Function enrichment and network analysis of the differentially expressed genes uncovered converging evidence on abnormal expression in key functional pathways, such as apoptosis, cell cycle and survival, and MAPK signaling in the SZ and SAD samples. By leveraging transcriptome profiles of normal human brain tissues across human development into adulthood, we showed that the differentially expressed genes converge on a sub-network mediated by CDC45 and the cell cycle, which would be disrupted by the 22q11.2 deletion during embryonic brain development, and another sub-network modulated by PRODH, which could contribute to disruption of brain function during adolescence. CONCLUSIONS This study has provided evidence for disruption of potential molecular events in SZ patient with 22q11.2 deletion and related our findings from in vitro neuronal cultures to functional perturbations that can occur during brain development in SZ.
Collapse
Affiliation(s)
- Mingyan Lin
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Anastasia Hrabovsky
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Jian Chen
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Benjamin R. Puliafito
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Stephanie R. Gilbert
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Neurology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| |
Collapse
|
20
|
Tetruashvily MM, Melson JW, Park JJ, Peng X, Boulanger LM. Expression and alternative splicing of classical and nonclassical MHCI genes in the hippocampus and neuromuscular junction. Mol Cell Neurosci 2016; 72:34-45. [PMID: 26802536 DOI: 10.1016/j.mcn.2016.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/01/2015] [Accepted: 01/15/2016] [Indexed: 01/25/2023] Open
Abstract
The major histocompatibility complex class I (MHCI) is a large gene family, with over 20 members in mouse. Some MHCIs are well-known for their critical roles in the immune response. Studies in mice which lack stable cell-surface expression of many MHCI proteins suggest that one or more MHCIs also play unexpected, essential roles in the establishment, function, and modification of neuronal synapses. However, there is little information about which genes mediate MHCI's effects in neurons. In this study, RT-PCR was used to simultaneously assess transcription of many MHCI genes in regions of the central and peripheral nervous system where MHCI has a known or suspected role. In the hippocampus, a part of the CNS where MHCI regulates synapse density, synaptic transmission, and plasticity, we found that more than a dozen MHCI genes are transcribed. Single-cell RT-PCR revealed that individual hippocampal neurons can express more than one MHCI gene, and that the MHCI gene expression profile of CA1 pyramidal neurons differs significantly from that of CA3 pyramidal neurons or granule cells of the dentate gyrus. MHCI gene expression was also assessed at the neuromuscular junction (NMJ), a part of the peripheral nervous system (PNS) where MHCI plays a role in developmental synapse elimination, aging-related synapse loss, and neuronal regeneration. Four MHCI genes are expressed at the NMJ at an age when synapse elimination is occurring in three different muscles. Several MHCI mRNA splice variants were detected in hippocampus, but not at the NMJ. Together, these results establish the first profile of MHCI gene expression at the developing NMJ, and demonstrate that MHCI gene expression is under tight spatial and temporal regulation in the nervous system. They also identify more than a dozen MHCIs that could play important roles in regulating synaptic transmission and plasticity in the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Mazell M Tetruashvily
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, United States; Rutgers Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08901, United States
| | - John W Melson
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, United States
| | - Joseph J Park
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, United States
| | - Xiaoyu Peng
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, United States; Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08544, United States
| | - Lisa M Boulanger
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, United States; Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08544, United States.
| |
Collapse
|
21
|
Mangold CA, Masser DR, Stanford DR, Bixler GV, Pisupati A, Giles CB, Wren JD, Ford MM, Sonntag WE, Freeman WM. CNS-wide Sexually Dimorphic Induction of the Major Histocompatibility Complex 1 Pathway With Aging. J Gerontol A Biol Sci Med Sci 2016; 72:16-29. [PMID: 26786204 DOI: 10.1093/gerona/glv232] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/09/2015] [Indexed: 01/01/2023] Open
Abstract
The major histocompatibility complex I (MHCI) pathway, which canonically functions in innate immune viral antigen presentation and detection, is functionally pleiotropic in the central nervous system (CNS). Alternative roles include developmental synapse pruning, regulation of synaptic plasticity, and inhibition of neuronal insulin signaling; all processes altered during brain aging. Upregulation of MHCI components with aging has been reported; however, no systematic examination of MHCI cellular localization, expression, and regulation across CNS regions, life span, and sexes has been reported. In the mouse, MHCI is expressed by neurons and microglia, and MHCI components and receptors (H2-K1, H2-D1, β2M, Lilrb3, Klra2, CD247) display markedly different expression profiles across the hippocampus, cortex, cerebellum, brainstem, and retina. MHCI components, receptors, associated inflammatory transcripts (IL1α, IL1β, IL6, TNFα), and TAP (transporter associated with antigen processing) components are induced with aging and to a greater degree in female than male mice across CNS regions. H2-K1 and H2-D1 expression is associated with differential CG and non-CG promoter methylation across CNS regions, ages, and between sexes, and concomitant increased expression of proinflammatory genes. Meta-analysis of human brain aging data also demonstrates age-related increases in MHCI. Induction of MHCI signaling could contribute to altered synapse regulation and impaired synaptic plasticity with aging.
Collapse
Affiliation(s)
- Colleen A Mangold
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey
| | - Dustin R Masser
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey.,Department of Physiology, University of Oklahoma Health Sciences Center.,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - David R Stanford
- Department of Physiology, University of Oklahoma Health Sciences Center.,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Georgina V Bixler
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey
| | - Aditya Pisupati
- MD/PhD Program, College of Medicine, Pennsylvania State University, Hershey
| | - Cory B Giles
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation
| | - Matthew M Ford
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Willard M Freeman
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey. .,Department of Physiology, University of Oklahoma Health Sciences Center.,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| |
Collapse
|
22
|
MHC class I limits hippocampal synapse density by inhibiting neuronal insulin receptor signaling. J Neurosci 2014; 34:11844-56. [PMID: 25164678 DOI: 10.1523/jneurosci.4642-12.2014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Proteins of the major histocompatibility complex class I (MHCI) negatively regulate synapse density in the developing vertebrate brain (Glynn et al., 2011; Elmer et al., 2013; Lee et al., 2014), but the underlying mechanisms remain largely unknown. Here we identify a novel MHCI signaling pathway that involves the inhibition of a known synapse-promoting factor, the insulin receptor. Dominant-negative insulin receptor constructs decrease synapse density in the developing Xenopus visual system (Chiu et al., 2008), and insulin receptor activation increases dendritic spine density in mouse hippocampal neurons in vitro (Lee et al., 2011). We find that genetically reducing cell surface MHCI levels increases synapse density selectively in regions of the hippocampus where insulin receptors are expressed, and occludes the neuronal insulin response by de-repressing insulin receptor signaling. Pharmacologically inhibiting insulin receptor signaling in MHCI-deficient animals rescues synapse density, identifying insulin receptor signaling as a critical mediator of the tonic inhibitory effects of endogenous MHCI on synapse number. Insulin receptors co-immunoprecipitate MHCI from hippocampal lysates, and MHCI unmasks a cytoplasmic epitope of the insulin receptor that mediates downstream signaling. These results identify an important role for an MHCI-insulin receptor signaling pathway in circuit patterning in the developing brain, and suggest that changes in MHCI expression could unexpectedly regulate neuronal insulin sensitivity in the aging and diseased brain.
Collapse
|
23
|
McAllister AK. Major histocompatibility complex I in brain development and schizophrenia. Biol Psychiatry 2014; 75:262-8. [PMID: 24199663 PMCID: PMC4354937 DOI: 10.1016/j.biopsych.2013.10.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/24/2013] [Accepted: 10/07/2013] [Indexed: 02/01/2023]
Abstract
Although the etiology of schizophrenia (SZ) remains unknown, it is increasingly clear that immune dysregulation plays a central role. Genome-wide association studies reproducibly indicate an association of SZ with immune genes within the major histocompatibility complex (MHC). Moreover, environmental factors that increase risk for SZ, such as maternal infection, alter peripheral immune responses as well as the expression of immune molecules in the brain. MHC class I (MHCI) molecules might mediate both genetic and environmental contributions to SZ through direct effects on brain development in addition to mediating immunity. MHCI molecules are expressed on neurons in the central nervous system throughout development and into adulthood, where they regulate many aspects of brain development, including neurite outgrowth, synapse formation and function, long-term and homeostatic plasticity, and activity-dependent synaptic refinement. This review summarizes our current understanding of MHCI expression and function in the developing brain as well as its involvement in maternal immune activation, from the perspective of how these roles for MHCI molecules might contribute to the pathogenesis of SZ.
Collapse
|
24
|
Lv D, Shi Q, Liu J, Zhang A, Miao F, He Y, Shen Y, Zhang J. The similar expression pattern of MHC class I molecules in human and mouse cerebellar cortex. Neurochem Res 2013; 39:180-6. [PMID: 24272393 DOI: 10.1007/s11064-013-1204-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 10/22/2013] [Accepted: 11/18/2013] [Indexed: 11/29/2022]
Abstract
The major histocompatibility complex (MHC) class I molecules are considered to be important in the immune system. However, the results reported in the past decade indicate that they also play important roles in the central nervous system. Here we examined the expression of MHC I and β2-microglobulin (β2m) in human and mouse cerebellar cortex. The results show that MHC I molecules are expressed both in human and mouse cerebellar cortex during brain development. The expression of H-2K(b)/D(b) is gradually increased with the development of mouse cerebellar cortex, but finally decreased to a very low level. Similarly, the expression of HLA-B/C genes is increased in developing human cerebellar cortex, but decreased after birth. The spatial and temporal expression of β2m overlaps mostly with that of HLA-B/C molecules, and they are co-expressed in Purkinje cells. Our findings provide a fundamental basis to reveal the functions of neuronal MHC class I molecules in the development of human cerebellum.
Collapse
Affiliation(s)
- Dan Lv
- Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu Province, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Salcedo E, Cruz NM, Ly X, Welander BA, Hanson K, Kronberg E, Restrepo D. A TAP1 null mutation leads to an enlarged olfactory bulb and supernumerary, ectopic olfactory glomeruli. Open Biol 2013; 3:130044. [PMID: 23697805 PMCID: PMC3866874 DOI: 10.1098/rsob.130044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Major histocompatibility class I (MHCI) molecules are well known for their immunological role in mediating tissue graft rejection. Recently, these molecules were discovered to be expressed in distinct neuronal subclasses, dispelling the long-held tenet that the uninjured brain is immune-privileged. Here, we show that MHCI molecules are expressed in the main olfactory bulb (MOB) of adult animals. Furthermore, we find that mice with diminished levels of MHCI expression have enlarged MOBs containing an increased number of small, morphologically abnormal and ectopically located P2 glomeruli. These findings suggest that MHCI molecules may play an important role in the proper formation of glomeruli in the bulb.
Collapse
Affiliation(s)
- Ernesto Salcedo
- Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Debnath M, Cannon DM, Venkatasubramanian G. Variation in the major histocompatibility complex [MHC] gene family in schizophrenia: associations and functional implications. Prog Neuropsychopharmacol Biol Psychiatry 2013; 42:49-62. [PMID: 22813842 DOI: 10.1016/j.pnpbp.2012.07.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/23/2012] [Accepted: 07/09/2012] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a chronic debilitating neuropsychiatric disorder with a complex genetic contribution. Although multiple genetic, immunological and environmental factors are known to contribute to schizophrenia susceptibility, the underlying neurobiological mechanism(s) is yet to be established. The immune system dysfunction theory of schizophrenia is experiencing a period of renewal due to a growth in evidence implicating components of the immune system in brain function and human behavior. Current evidence indicates that certain immune molecules such as Major Histocompatibility Complex (MHC) and cytokines, the key regulators of immunity and inflammation are directly involved in the neurobiological processes related to neurodevelopment, neuronal plasticity, learning, memory and behavior. However, the strongest support in favor of the immune hypothesis has recently emerged from on-going genome wide association studies advocating MHC region variants as major determinants of one's risk for developing schizophrenia. Further identification of the interacting partners and receptors of MHC molecules in the brain and their role in down-stream signaling pathways of neurotransmission have implicated these molecules as potential schizophrenia risk factors. More recently, combined brain imaging and genetic studies have revealed a relationship between genetic variations within the MHC region and neuromorphometric changes during schizophrenia. Furthermore, MHC molecules play a significant role in the immune-infective and neurodevelopmental pathogenetic pathways, currently hypothesized to contribute to the pathophysiology of schizophrenia. Herein, we review the immunological, genetic and expression studies assessing the role of the MHC in conferring risk for developing schizophrenia, we summarize and discuss the possible mechanisms involved, making note of the challenges to, and future directions of, immunogenetic research in schizophrenia.
Collapse
Affiliation(s)
- Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore-560029, India.
| | | | | |
Collapse
|
27
|
Zhang Q, Rahim MMA, Allan DSJ, Tu MM, Belanger S, Abou-Samra E, Ma J, Sekhon HS, Fairhead T, Zein HS, Carlyle JR, Anderson SK, Makrigiannis AP. Mouse Nkrp1-Clr gene cluster sequence and expression analyses reveal conservation of tissue-specific MHC-independent immunosurveillance. PLoS One 2012; 7:e50561. [PMID: 23226525 PMCID: PMC3514311 DOI: 10.1371/journal.pone.0050561] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/23/2012] [Indexed: 01/23/2023] Open
Abstract
The Nkrp1 (Klrb1)-Clr (Clec2) genes encode a receptor-ligand system utilized by NK cells as an MHC-independent immunosurveillance strategy for innate immune responses. The related Ly49 family of MHC-I receptors displays extreme allelic polymorphism and haplotype plasticity. In contrast, previous BAC-mapping and aCGH studies in the mouse suggest the neighboring and related Nkrp1-Clr cluster is evolutionarily stable. To definitively compare the relative evolutionary rate of Nkrp1-Clr vs. Ly49 gene clusters, the Nkrp1-Clr gene clusters from two Ly49 haplotype-disparate inbred mouse strains, BALB/c and 129S6, were sequenced. Both Nkrp1-Clr gene cluster sequences are highly similar to the C57BL/6 reference sequence, displaying the same gene numbers and order, complete pseudogenes, and gene fragments. The Nkrp1-Clr clusters contain a strikingly dissimilar proportion of repetitive elements compared to the Ly49 clusters, suggesting that certain elements may be partly responsible for the highly disparate Ly49 vs. Nkrp1 evolutionary rate. Focused allelic polymorphisms were found within the Nkrp1b/d (Klrb1b), Nkrp1c (Klrb1c), and Clr-c (Clec2f) genes, suggestive of possible immune selection. Cell-type specific transcription of Nkrp1-Clr genes in a large panel of tissues/organs was determined. Clr-b (Clec2d) and Clr-g (Clec2i) showed wide expression, while other Clr genes showed more tissue-specific expression patterns. In situ hybridization revealed specific expression of various members of the Clr family in leukocytes/hematopoietic cells of immune organs, various tissue-restricted epithelial cells (including intestinal, kidney tubular, lung, and corneal progenitor epithelial cells), as well as myocytes. In summary, the Nkrp1-Clr gene cluster appears to evolve more slowly relative to the related Ly49 cluster, and likely regulates innate immunosurveillance in a tissue-specific manner.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Mir Munir A. Rahim
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - David S. J. Allan
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Megan M. Tu
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Simon Belanger
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Elias Abou-Samra
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jaehun Ma
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Harman S. Sekhon
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Todd Fairhead
- Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Haggag S. Zein
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Department of Genetics, Cairo University, Giza, Egypt
| | - James R. Carlyle
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Stephen K. Anderson
- Basic Science Program, SAIC-Frederick Inc., Laboratory of Experimental Immunology, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Andrew P. Makrigiannis
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
28
|
Chacon MA, Boulanger LM. MHC class I protein is expressed by neurons and neural progenitors in mid-gestation mouse brain. Mol Cell Neurosci 2012; 52:117-27. [PMID: 23147111 DOI: 10.1016/j.mcn.2012.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 10/09/2012] [Accepted: 11/02/2012] [Indexed: 02/04/2023] Open
Abstract
Proteins of the major histocompatibility complex class I (MHCI) are known for their role in the vertebrate adaptive immune response, and are required for normal postnatal brain development and plasticity. However, it remains unknown if MHCI proteins are present in the mammalian brain before birth. Here, we show that MHCI proteins are widely expressed in the developing mouse central nervous system at mid-gestation (E9.5-10.5). MHCI is strongly expressed in several regions of the prenatal brain, including the neuroepithelium and olfactory placode. MHCI is expressed by neural progenitors at these ages, as identified by co-expression in cells positive for neuron-specific class III β-tubulin (Tuj1) or for Pax6, a marker of neural progenitors in the dorsal neuroepithelium. MHCI is also co-expressed with nestin, a marker of neural stem/progenitor cells, in olfactory placode, but the co-localization is less extensive in other regions. MHCI is detected in the small population of post-mitotic neurons that are present at this early stage of brain development, as identified by co-expression in cells positive for neuronal microtubule-associated protein-2 (MAP2). Thus MHCI protein is expressed during the earliest stages of neuronal differentiation in the mammalian brain. MHCI expression in neurons and neural progenitors at mid-gestation, prior to the maturation of the adaptive immune system, is consistent with MHCI performing non-immune functions in prenatal brain development. These results raise the possibility that disruption of the levels and/or patterns of MHCI expression in the prenatal brain could contribute to the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Marcelo A Chacon
- Department of Molecular Biology and Princeton Neuroscience Institute, Princeton University, 123 Lewis Thomas Laboratories, Washington Road, Princeton, NJ 08544, USA
| | | |
Collapse
|
29
|
Elmer BM, McAllister AK. Major histocompatibility complex class I proteins in brain development and plasticity. Trends Neurosci 2012; 35:660-70. [PMID: 22939644 DOI: 10.1016/j.tins.2012.08.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 06/27/2012] [Accepted: 08/03/2012] [Indexed: 10/28/2022]
Abstract
Proper development of the central nervous system (CNS) requires the establishment of appropriate connections between neurons. Recent work suggests that this process is controlled by a balance between synaptogenic molecules and proteins that negatively regulate synapse formation and plasticity. Surprisingly, many of these newly identified synapse-limiting molecules are classic 'immune' proteins. In particular, major histocompatibility complex class I (MHCI) molecules regulate neurite outgrowth, the establishment and function of cortical connections, activity-dependent refinement in the visual system, and long-term and homeostatic plasticity. This review summarizes our current understanding of MHCI expression and function in the CNS, as well as the potential mechanisms used by MHCI to regulate brain development and plasticity.
Collapse
Affiliation(s)
- Bradford M Elmer
- Center for Neuroscience, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | | |
Collapse
|
30
|
Zhang X, Reed EF. HLA class I: an unexpected role in integrin β4 signaling in endothelial cells. Hum Immunol 2012; 73:1239-44. [PMID: 22789625 DOI: 10.1016/j.humimm.2012.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 06/11/2012] [Accepted: 06/29/2012] [Indexed: 12/31/2022]
Abstract
The production of anti-donor antibodies to HLA class I and class II antigens following transplantation is associated with development of transplant vasculopathy and graft loss. Antibodies against HLA class I (HLA-I) molecules are thought to contribute to transplant vasculopathy by triggering signals that elicit the activation and proliferation of endothelial cells. The proximal molecular events that regulate HLA-I dependent signal transduction are not well understood. We demonstrated a mutual dependency between HLA-I and integrin β4 to stimulate signal transduction and cell proliferation. Similarly, we found that integrin β4-mediated cell migration was dependent upon its interactions with HLA-I molecules. Since integrin β4 has been implicated in angiogenesis and tumor formation, associations between integrin β4 and HLA-I may play an important role in cancer. Further characterization of interactions between HLA-I and integrin β4 may lead to the development of therapeutic strategies for the treatment and prevention of chronic allograft rejection and cancer.
Collapse
Affiliation(s)
- Xiaohai Zhang
- UCLA Immunogenetics Center, Department of Pathology, David Geffen School of Medicine, University of California, 1000 Veteran Ave., Los Angeles, CA 90095, United States.
| | | |
Collapse
|
31
|
VanGuilder Starkey HD, Van Kirk CA, Bixler GV, Imperio CG, Kale VP, Serfass JM, Farley JA, Yan H, Warrington JP, Han S, Mitschelen M, Sonntag WE, Freeman WM. Neuroglial expression of the MHCI pathway and PirB receptor is upregulated in the hippocampus with advanced aging. J Mol Neurosci 2012; 48:111-26. [PMID: 22562814 DOI: 10.1007/s12031-012-9783-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/16/2012] [Indexed: 12/31/2022]
Abstract
The hippocampus undergoes changes with aging that impact neuronal function, such as synapse loss and altered neurotransmitter release. Nearly half of the aged population also develops deficits in spatial learning and memory. To identify age-related hippocampal changes that may contribute to cognitive decline, transcriptomic analysis of synaptosome preparations from adult (12 months) and aged (28 months) Fischer 344-Brown Norway rats assessed for spatial learning and memory was performed. Bioinformatic analysis identified the MHCI pathway as significantly upregulated with aging. Age-related increases in mRNAs encoding the MHCI genes RT1-A1, RT1-A2, and RT1-A3 were confirmed by qPCR in synaptosomes and in CA1 and CA3 dissections. Elevated levels of the MHCI cofactor (B2m), antigen-loading components (Tap1, Tap2, Tapbp), and two known MHCI receptors (PirB, Klra2) were also confirmed. Protein expression of MHCI was elevated with aging in synaptosomes, CA1, and DG, while PirB protein expression was induced in both CA1 and DG. MHCI expression was localized to microglia and neuronal excitatory postsynaptic densities, and PirB was localized to neuronal somata, axons, and dendrites. Induction of the MHCI antigen processing and presentation pathway in hippocampal neurons and glia may contribute to age-related hippocampal dysfunction by increasing neuroimmune signaling or altering synaptic homeostasis.
Collapse
|
32
|
Major histocompatibility complex class I molecules modulate embryonic neuritogenesis and neuronal polarization. J Neuroimmunol 2012; 247:1-8. [PMID: 22503373 DOI: 10.1016/j.jneuroim.2012.03.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/08/2012] [Accepted: 03/12/2012] [Indexed: 11/20/2022]
Abstract
We studied cultured hippocampal neurons from embryonic wildtype, major histocompatibility complex class I (MHCI) heavy chain-deficient (K(b)D(b)-/-) and NSE-D(b) (which have elevated neuronal MHCI expression) C57BL/6 mice. K(b)D(b)-/- neurons displayed slower neuritogenesis and establishment of polarity, while NSE-D(b) neurons had faster neurite outgrowth, more primary neurites, and tended to have accelerated polarization. Additional studies with ß2M-/- neurons, exogenous ß2M, and a self-MHCI monomer suggest that free heavy chain cis interactions with other surface molecules can promote neuritogenesis while tripartite MHCI interactions with classical MHCI receptors can inhibit axon outgrowth. Together with the results of others, MHCI appears to differentially modulate neuritogenesis and synaptogenesis.
Collapse
|
33
|
Yewdell JW. DRiPs solidify: progress in understanding endogenous MHC class I antigen processing. Trends Immunol 2011; 32:548-58. [PMID: 21962745 DOI: 10.1016/j.it.2011.08.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 07/27/2011] [Accepted: 08/01/2011] [Indexed: 12/19/2022]
Abstract
Defective ribosomal products (DRiPs) are a subset of rapidly degraded polypeptides that provide peptide ligands for major histocompatibility complex (MHC) class I molecules. Here, recent progress in understanding DRiP biogenesis is reviewed. These findings place DRiPs at the center of the MHC class I antigen processing pathway, linking immunosurveillance of viruses and tumors to mechanisms of specialized translation and cellular compartmentalization. DRiPs enable the immune system to rapidly detect alterations in cellular gene expression with great sensitivity.
Collapse
|
34
|
Cullheim S, Thams S. Classic major histocompatibility complex class I molecules: new actors at the neuromuscular junction. Neuroscientist 2011; 16:600-7. [PMID: 21239728 DOI: 10.1177/1073858410381534] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The presence and function of immune molecules in the central nervous system (CNS) have been under debate for a long time. There is mounting evidence that molecules fundamental for immune function are indeed expressed by both neurons and glia and that such molecules may have important nonimmunological function for the organization and stability of synaptic connections. Here, we present data showing that the classic form of major histocompatibility complex (MHC) class I molecules is expressed in spinal motoneurons, in particular in their axons and presynaptically at their synapses with skeletal muscles, the neuromuscular junctions (NMJs). The expression is strongly increased after axon lesion in the peripheral nerve. In the absence of classic MHC I, the organization of NMJs is disturbed with NMJs in higher numbers than normal, thereby equipping single muscle fibers with multiple NMJs. It is suggested that these effects are mediated by the classic MHC class I in the motor axons, possibly through effects mediated by the peripherally myelinating Schwann cells, which express receptors for classic MHC class I. The presence of immune molecules normally used by other cells for antigen presentation in peripheral motor axons may have implications for the onset of specific motoneuron disease.
Collapse
Affiliation(s)
- Staffan Cullheim
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
35
|
Wu ZP, Bilousova T, Escande-Beillard N, Dang H, Hsieh T, Tian J, Kaufman DL. Major histocompatibility complex class I-mediated inhibition of neurite outgrowth from peripheral nerves. Immunol Lett 2011; 135:118-23. [PMID: 20974178 PMCID: PMC5776043 DOI: 10.1016/j.imlet.2010.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 10/14/2010] [Accepted: 10/17/2010] [Indexed: 10/18/2022]
Abstract
Studies of mice deficient in classical major histocompatability complex class I (MHCI) revealed that MHCI plays an important role in neurodevelopment in the central nervous system. We previously studied the effects of recombinant MHCI molecules on wildtype retina explants and observed that MHCI can inhibit retina neurite outgrowth, with self-MHCI molecules having greater inhibitory effect than non-self MHCI molecules. Here, we examined classical MHCI's effects on axon outgrowth from neurons of the peripheral nervous system (PNS). We used the embryonic dorsal root ganglia (DRG) explant model since their neurons express MHCI and because DRG explants have been widely used to assess the effects of molecules on axonal outgrowth from PNS neurons. We observed that picomolar levels of a recombinant self-MHCI molecule, but not non-self MHCI molecules, inhibited axon outgrowth from DRG explants. This differential sensitivity to self- vs. non-self MHCI suggests that early in development, self-MHCI may "educate" PNS neurons to express appropriate MHCI receptors, as occurs during natural killer cell development. Furthermore, we observed that a MHCI tetramer stained embryonic DRG neurons, indicating the expression of classical MHCI receptors. These results suggest that MHCI and MHCI receptors play roles during early stages of PNS development and may provide new targets of therapeutic strategies to promote neuronal outgrowth after PNS injury.
Collapse
Affiliation(s)
- Zhongqi-Phyllis Wu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Tina Bilousova
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Nathalie Escande-Beillard
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Hoa Dang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Terry Hsieh
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Jide Tian
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Daniel L. Kaufman
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| |
Collapse
|
36
|
Wu ZP, Washburn L, Bilousova TV, Boudzinskaia M, Escande-Beillard N, Querubin J, Dang H, Xie CW, Tian J, Kaufman DL. Enhanced neuronal expression of major histocompatibility complex class I leads to aberrations in neurodevelopment and neurorepair. J Neuroimmunol 2011; 232:8-16. [PMID: 20950866 PMCID: PMC5776042 DOI: 10.1016/j.jneuroim.2010.09.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 09/15/2010] [Indexed: 01/22/2023]
Abstract
Mice deficient in classical major histocompatibility complex class I (MHCI) have aberrations in neurodevelopment. The consequences of upregulated neuronal MHCI expression have not been examined. We found that transgenic C57Bl/6 mice that are engineered to express higher levels of self-D(b) on their CNS neurons have alterations in their hippocampal morphology and retinogeniculate projections, as well as impaired neurorepair responses. Thus, enhanced neuronal classical MHCI expression can lead to aberrations in neural circuitry and neurorepair. These findings complement a growing body of knowledge concerning the neurobiological activities of MHCI and may have potential clinical relevance.
Collapse
Affiliation(s)
- Zhongqi-Phyllis Wu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Lorraine Washburn
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Tina V. Bilousova
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Maia Boudzinskaia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Nathalie Escande-Beillard
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Jyes Querubin
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Hoa Dang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Cui-Wei Xie
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Jide Tian
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Daniel L. Kaufman
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| |
Collapse
|
37
|
Fourgeaud L, Boulanger LM. Role of immune molecules in the establishment and plasticity of glutamatergic synapses. Eur J Neurosci 2011; 32:207-17. [PMID: 20946111 DOI: 10.1111/j.1460-9568.2010.07342.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An increasing number of studies support an unexpected role for immune molecules in regulating healthy brain functions during development and in adulthood. Here we review the roles of specific immune molecules (including cytokines, components of the complement cascade, and members of the major histocompatibility complex class I family and their receptors) in the formation and plasticity of glutamatergic synapses. These findings add a new dimension to our understanding of neural-immune interactions, and suggest novel molecular mechanisms that may underlie the modification of glutamatergic synapses in both normal and pathological states.
Collapse
Affiliation(s)
- Lawrence Fourgeaud
- Molecular Neurobiology Laboratories (MNL-L), The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | |
Collapse
|
38
|
MHC class I modulates NMDA receptor function and AMPA receptor trafficking. Proc Natl Acad Sci U S A 2010; 107:22278-83. [PMID: 21135233 DOI: 10.1073/pnas.0914064107] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Proteins of the major histocompatibility complex class I (MHCI) are known for their role in immunity and have recently been implicated in long-term plasticity of excitatory synaptic transmission. However, the mechanisms by which MHCI influences synaptic plasticity remain unknown. Here we show that endogenous MHCI regulates synaptic responses mediated by NMDA-type glutamate receptors (NMDARs) in the mammalian central nervous system (CNS). The AMPA/NMDA ratio is decreased at MHCI-deficient hippocampal synapses, reflecting an increase in NMDAR-mediated currents. This enhanced NMDAR response is not associated with changes in the levels, subunit composition, or gross subcellular distribution of NMDARs. Increased NMDAR-mediated currents in MHCI-deficient neurons are associated with characteristic changes in AMPA receptor trafficking in response to NMDAR activation. Thus, endogenous MHCI tonically inhibits NMDAR function and controls downstream NMDAR-induced AMPA receptor trafficking during the expression of plasticity.
Collapse
|
39
|
Zhang X, Rozengurt E, Reed EF. HLA class I molecules partner with integrin β4 to stimulate endothelial cell proliferation and migration. Sci Signal 2010; 3:ra85. [PMID: 21098729 PMCID: PMC3878299 DOI: 10.1126/scisignal.2001158] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Among transplant recipients, those who produce antibodies against the donor's human leukocyte antigens (HLAs) are at higher risk for antibody-mediated rejection and transplant vasculopathy, which is a progressive, vasculo-occlusive disease that results in ischemic injury and deterioration of organ function. Antibodies against HLA class I (HLA-I) molecules are thought to contribute to transplant vasculopathy by triggering signals that elicit the activation and proliferation of endothelial cells. Here, we demonstrate a molecular association between HLA-I and the integrin β(4) subunit after the stimulation of endothelial cells with HLA-I-specific antibodies. Knockdown of integrin β(4) in these cells abrogated the ability of HLA-I to stimulate the phosphorylation of the kinases Akt, extracellular signal-regulated kinase (ERK), and Src, as well as cellular proliferation. Similarly, reducing the abundance of HLA-I suppressed integrin β(4)-mediated phosphorylation of ERK and the migration of endothelial cells on laminin-5, a component of the extracellular matrix. These results indicate a mutual dependency between HLA-I and the integrin β(4) subunit to stimulate the proliferation and migration of endothelial cells, which may be important in promoting transplant vasculopathy and tumor angiogenesis.
Collapse
Affiliation(s)
- Xiaohai Zhang
- Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, Center for Ulcer Research and Education, Digestive Diseases Research Center, David Geffen School of Medicine and Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Elaine F. Reed
- Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Garay PA, McAllister AK. Novel roles for immune molecules in neural development: implications for neurodevelopmental disorders. Front Synaptic Neurosci 2010; 2:136. [PMID: 21423522 PMCID: PMC3059681 DOI: 10.3389/fnsyn.2010.00136] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 08/03/2010] [Indexed: 11/18/2022] Open
Abstract
Although the brain has classically been considered “immune-privileged”, current research suggests an extensive communication between the immune and nervous systems in both health and disease. Recent studies demonstrate that immune molecules are present at the right place and time to modulate the development and function of the healthy and diseased central nervous system (CNS). Indeed, immune molecules play integral roles in the CNS throughout neural development, including affecting neurogenesis, neuronal migration, axon guidance, synapse formation, activity-dependent refinement of circuits, and synaptic plasticity. Moreover, the roles of individual immune molecules in the nervous system may change over development. This review focuses on the effects of immune molecules on neuronal connections in the mammalian central nervous system – specifically the roles for MHCI and its receptors, complement, and cytokines on the function, refinement, and plasticity of geniculate, cortical and hippocampal synapses, and their relationship to neurodevelopmental disorders. These functions for immune molecules during neural development suggest that they could also mediate pathological responses to chronic elevations of cytokines in neurodevelopmental disorders, including autism spectrum disorders (ASD) and schizophrenia.
Collapse
Affiliation(s)
- Paula A Garay
- Laboratory of Dr. A.K. McAllister, Department of Neurobiology, Physiology, and Behavior, Center for Neuroscience, University of California Davis, CA, USA
| | | |
Collapse
|
41
|
McDole JR, Danzer SC, Pun RYK, Chen Y, Johnson HL, Pirko I, Johnson AJ. Rapid formation of extended processes and engagement of Theiler's virus-infected neurons by CNS-infiltrating CD8 T cells. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1823-33. [PMID: 20813972 DOI: 10.2353/ajpath.2010.100231] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A fundamental question in neuroimmunology is the extent to which CD8 T cells actively engage virus-infected neurons. In the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis, an effective central nervous system (CNS)-infiltrating antiviral CD8 T cell response offers protection from this demyelinating disease. However, the specific CNS cell types engaged by these protective CD8 T cells in TMEV-resistant strains remains unknown. We used confocal microscopy to visualize the morphology, migration, and specific cellular interactions between adoptively transferred CD8 T cells and specific CNS cell types. Adoptively transferred GFP+ CD8+ splenocytes migrated to the brain and became 93% specific for the immunodominant virus epitope D(b):VP2(121-130). These CD8 T cells also polarized T cell receptor, CD8 protein, and granzyme B toward target neurons. Furthermore, we observed CD8 T cells forming cytoplasmic processes up to 45 μm in length. Using live tissue imaging, we determined that these T cell-extended processes (TCEPs) could be rapidly formed and were associated with migratory behavior through CNS tissues. These studies provide evidence that antiviral CD8 T cells have the capacity to engage virus-infected neurons in vivo and are the first to document and measure the rapid formation of TCEPs on these brain-infiltrating lymphocytes using live tissue imaging.
Collapse
Affiliation(s)
- Jeremiah R McDole
- Departments of Neurology,University of Cincinnati College of Medicine , Cincinnati, OH 45267, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Escande-Beillard N, Washburn L, Zekzer D, Wu ZP, Eitan S, Ivkovic S, Lu Y, Dang H, Middleton B, Bilousova TV, Yoshimura Y, Evans CJ, Joyce S, Tian J, Kaufman DL. Neurons preferentially respond to self-MHC class I allele products regardless of peptide presented. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:816-23. [PMID: 20018625 PMCID: PMC2997386 DOI: 10.4049/jimmunol.0902159] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Studies of mice lacking MHC class I (MHC I)-associated proteins have demonstrated a role for MHC I in neurodevelopment. A central question arising from these observations is whether neuronal recognition of MHC I has specificity for the MHC I allele product and the peptide presented. Using a well-established embryonic retina explant system, we observed that picomolar levels of a recombinant self-MHC I molecule inhibited neurite outgrowth. We then assessed the neurobiological activity of a panel of recombinant soluble MHC Is, consisting of different MHC I heavy chains with a defined self- or nonself-peptide presented, on cultured embryonic retinas from mice with different MHC I haplotypes. We observed that self-MHC I allele products had greater inhibitory neuroactivity than nonself-MHC I molecules, regardless of the nature of the peptide presented, a pattern akin to MHC I recognition by some innate immune system receptors. However, self-MHC I molecules had no effect on retinas from MHC I-deficient mice. These observations suggest that neuronal recognition of MHC I may be coordinated with the inherited MHC I alleles, as occurs in the innate immune system. Consistent with this notion, we show that MHC I and MHC I receptors are coexpressed by precursor cells at the earliest stages of retina development, which could enable such coordination.
Collapse
Affiliation(s)
- Nathalie Escande-Beillard
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Classical major histocompatibility complex class I molecules in motoneurons: new actors at the neuromuscular junction. J Neurosci 2009; 29:13503-15. [PMID: 19864563 DOI: 10.1523/jneurosci.0981-09.2009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Major histocompatibility complex (MHC) class I molecules have fundamental functions in the immune system. Recent studies have suggested that these molecules may also have non-immune functions in the nervous system, in particular related to synaptic function and plasticity. Because adult motoneurons express mRNAs for MHC class I molecules, we have examined their subcellular expression pattern in vivo and their role for the synaptic connectivity of these neurons. We observed immunoreactivity for classical MHC class I (Ia) protein in motoneuron somata, but the predominant expression was found in axons and presynaptically at neuromuscular junctions (NMJs). Peripheral nerve lesion induced a strong increase of motoneuron MHC class Ia (H2-K(b)/D(b)) mRNA, indicating a role for MHC class Ia molecules during regeneration. Accordingly, there was an accumulation of MHC class Ia proteins at the cut ends and in growth cones of motor axons after lesion. In K(b-/-)D(b-/-) mice (lacking MHC class Ia molecules), the time course for recovery of grip ability in reinnervated muscles was significantly delayed. Muscles from K(b-/-)D(b-/-) mice displayed an increased density and a disturbed distribution of NMJs and fewer terminal Schwann cells/NMJ compared with wild-type mice. A population of Schwann cells in sciatic nerves expressed the paired Ig receptor B, which binds to MHC class I molecules. These results provide the first evidence that neuronal MHC class Ia molecules are present in motor axons, that they are important for organization of NMJs and motor recovery after nerve lesion, and that their actions may be mediated via Schwann cells.
Collapse
|
44
|
Abstract
Many proteins first identified in the immune system are also expressed in the developing and adult nervous system. Unexpectedly, recent studies reveal that a number of these proteins, in addition to their immunological roles, are essential for the establishment, function, and modification of synaptic connections. These include proinflammatory cytokines (e.g., TNFalpha, IL-6), proteins of the innate immune system (e.g., complement C1q and C3, pentraxins, Dscam), members of the major histocompatibility complex class I (MHCI) family, and MHCI-binding immunoreceptors and their components (e.g., PIRB, Ly49, DAP12, CD3zeta). Understanding how these proteins function in neurons will clarify the molecular basis of fundamental events in brain development and plasticity and may add a new dimension to our understanding of neural-immune interactions in health and disease.
Collapse
|
45
|
Abstract
For the nervous system to translate experience into memory and behavior, lasting structural change at synapses must occur. This requirement is clearly evident during critical periods of activity-dependent neural development, and accumulating evidence has established a surprising role for the major histocompatibility complex class I (MHCI) proteins in this process.
Collapse
Affiliation(s)
- Carla J Shatz
- Bio-X and Departments of Biology and Neurobiology, James H. Clark Center, 318 Campus Drive W1.1, Stanford University, Stanford CA 94305-5437, USA.
| |
Collapse
|
46
|
H2-K(b) and H2-D(b) regulate cerebellar long-term depression and limit motor learning. Proc Natl Acad Sci U S A 2009; 106:6784-9. [PMID: 19346486 DOI: 10.1073/pnas.0902018106] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
There are more than 50 class I MHC (MHCI) molecules in the mouse genome, some of which are now known to be expressed in neurons; however, the role of classical MHCI molecules in synaptic plasticity is unknown. We report that the classical MHCI molecules, H2-K(b) and H2-D(b), are co-expressed by Purkinje cells (PCs). In the cerebellum of mice deficient for both H2-K(b) and H2-D(b) (K(b)D(b-/-)), there is a lower threshold for induction of long-term depression (LTD) at parallel fiber to PC synapses. This change may be a result of additional glutamate release observed at K(b)D(b-/-) CF to PC synapses, which are thought to "train" the cerebellar circuit. A behavioral correlate of cerebellar LTD is motor learning; acquisition and retention of a Rotarod behavioral task is significantly better in K(b)D(b-/-) mice than in WT cohorts. These physiological and behavioral phenotypes in K(b)D(b-/-) mice reveal a surprising role for classical MHCI molecules in synaptic plasticity and motor learning.
Collapse
|