MHC class I H2-Kb negatively regulates neural progenitor cell proliferation by inhibiting FGFR signaling

PLoS Biology - Tập 19 Số 6 - Trang e3001311
Karin Lin1,2, Gregor Bieri1, Géraldine Gontier1, Sören Müller3, Lucas K. Smith4,1, Cedric E. Snethlage1, Charles White1,5, Sun Kim6, Saul Villeda4,1,7,5,2,8
1Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America.
2Neuroscience Graduate Program, University of California San Francisco, San Francisco, California, United States of America
3Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
4Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, California, United States of America
5Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, California, United States of America
6Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
7Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
8The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, San Francisco, California, United States of America

Tóm tắt

Proteins of the major histocompatibility complex class I (MHC I), predominantly known for antigen presentation in the immune system, have recently been shown to be necessary for developmental neural refinement and adult synaptic plasticity. However, their roles in nonneuronal cell populations in the brain remain largely unexplored. Here, we identify classical MHC I molecule H2-Kb as a negative regulator of proliferation in neural stem and progenitor cells (NSPCs). Using genetic knockout mouse models and in vivo viral-mediated RNA interference (RNAi) and overexpression, we delineate a role for H2-Kb in negatively regulating NSPC proliferation and adult hippocampal neurogenesis. Transcriptomic analysis of H2-Kb knockout NSPCs, in combination with in vitro RNAi, overexpression, and pharmacological approaches, further revealed that H2-Kb inhibits cell proliferation by dampening signaling pathways downstream of fibroblast growth factor receptor 1 (Fgfr1). These findings identify H2-Kb as a critical regulator of cell proliferation through the modulation of growth factor signaling.

Từ khóa


Tài liệu tham khảo

J Neefjes, 2011, Towards a systems understanding of MHC class I and MHC class II antigen presentation, Nat Rev Immunol, 11, 823, 10.1038/nri3084

PA Garay, 2010, Novel roles for immune molecules in neural development: implications for neurodevelopmental disorders, Front Synaptic Neurosci, 2, 136, 10.3389/fnsyn.2010.00136

CJ Shatz, 2009, MHC class I: an unexpected role in neuronal plasticity, Neuron, 64, 40, 10.1016/j.neuron.2009.09.044

BE Deverman, 2009, Cytokines and CNS development, Neuron, 64, 61, 10.1016/j.neuron.2009.09.002

B Stevens, 2007, The classical complement cascade mediates CNS synapse elimination, Cell, 131, 1164, 10.1016/j.cell.2007.10.036

TJ Dixon-Salazar, 2014, MHC class I limits hippocampal synapse density by inhibiting neuronal insulin receptor signaling, J Neurosci, 34, 11844, 10.1523/JNEUROSCI.4642-12.2014

CA Goddard, 2007, Regulation of CNS synapses by neuronal MHC class I, Proc Natl Acad Sci U S A, 104, 6828, 10.1073/pnas.0702023104

MW Glynn, 2011, MHCI negatively regulates synapse density during the establishment of cortical connections, Nat Neurosci, 14, 442, 10.1038/nn.2764

A Datwani, 2009, Classical MHCI molecules regulate retinogeniculate refinement and limit ocular dominance plasticity, Neuron, 64, 463, 10.1016/j.neuron.2009.10.015

GS Huh, 2000, Functional requirement for class I MHC in CNS development and plasticity, Science, 290, 2155, 10.1126/science.290.5499.2155

Z-P Wu, 2011, Major histocompatibility complex class I-mediated inhibition of neurite outgrowth from peripheral nerves, Immunol Lett, 135, 118, 10.1016/j.imlet.2010.10.011

AM Bond, 2015, Adult Mammalian Neural Stem Cells and Neurogenesis: Five Decades Later, Cell Stem Cell, 17, 385, 10.1016/j.stem.2015.09.003

G Kempermann, 2018, Human Adult Neurogenesis: Evidence and Remaining Questions, Cell Stem Cell, 23, 25, 10.1016/j.stem.2018.04.004

KM Christian, 2014, Functions and dysfunctions of adult hippocampal neurogenesis, Annu Rev Neurosci, 37, 243, 10.1146/annurev-neuro-071013-014134

R Favaro, 2009, Hippocampal development and neural stem cell maintenance require Sox2-dependent regulation of Shh, Nat Neurosci, 12, 1248, 10.1038/nn.2397

J-H Paik, 2009, FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis, Cell Stem Cell, 5, 540, 10.1016/j.stem.2009.09.013

G Gontier, 2018, Tet2 Rescues Age-Related Regenerative Decline and Enhances Cognitive Function in the Adult Mouse Brain, Cell Rep, 22, 1974, 10.1016/j.celrep.2018.02.001

Q Qu, 2010, Orphan nuclear receptor TLX activates Wnt/beta-catenin signalling to stimulate neural stem cell proliferation and self-renewal, Nat Cell Biol, 12, 1

SA Villeda, 2011, The ageing systemic milieu negatively regulates neurogenesis and cognitive function, Nature, 477, 90, 10.1038/nature10357

AM Horowitz, 2020, Blood factors transfer beneficial effects of exercise on neurogenesis and cognition to the aged brain, Science, 369, 167, 10.1126/science.aaw2622

CW White, 2020, Age-related loss of neural stem cell O-GlcNAc promotes a glial fate switch through STAT3 activation, Proc Natl Acad Sci U S A, 24, 202007439

A Borsini, 2015, The role of inflammatory cytokines as key modulators of neurogenesis, Trends Neurosci, 38, 145, 10.1016/j.tins.2014.12.006

A Rolls, 2007, Toll-like receptors modulate adult hippocampal neurogenesis, Nat Cell Biol, 9, 1081, 10.1038/ncb1629

Y Ziv, 2006, Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood, Nat Neurosci, 9, 268, 10.1038/nn1629

LK Smith, The aged hematopoietic system promotes hippocampal-dependent cognitive decline, Aging Cell. 2020, 19, e51481

LK Smith, 2015, β2-microglobulin is a systemic pro-aging factor that impairs cognitive function and neurogenesis, Nat Med, 21, 932, 10.1038/nm.3898

J Shin, 2015, Single-Cell RNA-Seq with Waterfall Reveals Molecular Cascades underlying Adult Neurogenesis, Cell Stem Cell, 17, 360, 10.1016/j.stem.2015.07.013

H Babu, 2011, A protocol for isolation and enriched monolayer cultivation of neural precursor cells from mouse dentate gyrus, Front Neurosci, 5, 89, 10.3389/fnins.2011.00089

I Tirosh, 2016, Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma, Nature, 539, 309, 10.1038/nature20123

FP Varodayan, 2009, Seizures increase cell proliferation in the dentate gyrus by shortening progenitor cell-cycle length, Epilepsia, 50, 2638, 10.1111/j.1528-1167.2009.02244.x

G Ponti, 2013, Cell cycle and lineage progression of neural progenitors in the ventricular-subventricular zones of adult mice, Proc Natl Acad Sci U S A, 110, E1045, 10.1073/pnas.1219563110

HD VanGuilder Starkey, 2012, Neuroglial expression of the MHCI pathway and PirB receptor is upregulated in the hippocampus with advanced aging, J Mol Neurosci, 48, 111, 10.1007/s12031-012-9783-8

JD Adelson, 2012, Neuroprotection from stroke in the absence of MHCI or PirB, Neuron, 73, 1100, 10.1016/j.neuron.2012.01.020

MM Tetruashvily, 2016, Boulanger LM. MHCI promotes developmental synapse elimination and aging-related synapse loss at the vertebrate neuromuscular junction, Brain Behav Immun, 56, 197, 10.1016/j.bbi.2016.01.008

H Lee, 2014, Synapse elimination and learning rules co-regulated by MHC class I H2-D(b.), Nature

K Jin, 2003, Neurogenesis and aging: FGF-2 and HB-EGF restore neurogenesis in hippocampus and subventricular zone of aged mice, Aging Cell, 2, 175, 10.1046/j.1474-9728.2003.00046.x

W Kang, 2015, Hébert JMFGF, Signaling I. Necessary for Neurogenesis in Young Mice and Sufficient to Reverse Its Decline in Old Mice, J Neurosci, 35, 10217, 10.1523/JNEUROSCI.1469-15.2015

ME Woodbury, 2014, Fibroblast growth factor-2 signaling in neurogenesis and neurodegeneration, J Neuroimmune Pharmacol, 9, 92, 10.1007/s11481-013-9501-5

G-L Ming, 2011, Adult neurogenesis in the mammalian brain: significant answers and significant questions, Neuron, 70, 687, 10.1016/j.neuron.2011.05.001

X Fan, 2017, Mechanisms of Hippocampal Aging and the Potential for Rejuvenation, Annu Rev Neurosci, 40, 251, 10.1146/annurev-neuro-072116-031357

International Schizophrenia Consortium, 2009, Common polygenic variation contributes to risk of schizophrenia and bipolar disorder, Nature, 460, 748, 10.1038/nature08185

WR Jeck, 2012, Review: a meta-analysis of GWAS and age-associated diseases, Aging Cell, 11, 727, 10.1111/j.1474-9726.2012.00871.x

J Shi, 2009, Common variants on chromosome 6p22.1 are associated with schizophrenia, Nature, 460, 753, 10.1038/nature08192

H Stefansson, 2009, Common variants conferring risk of schizophrenia, Nature, 460, 744, 10.1038/nature08186

RS Nowakowski, 1989, Bromodeoxyuridine immunohistochemical determination of the lengths of the cell cycle and the DNA-synthetic phase for an anatomically defined population, J Neurocytol, 18, 311, 10.1007/BF01190834

JJ Trombetta, 2014, Preparation of Single-Cell RNA-Seq Libraries for Next Generation Sequencing, Curr Protoc Mol Biol, 107, 4.22.1, 10.1002/0471142727.mb0422s107

D Kim, 2015, HISAT: a fast spliced aligner with low memory requirements, Nat Methods, 12, 357, 10.1038/nmeth.3317

Y Liao, 2014, featureCounts: an efficient general purpose program for assigning sequence reads to genomic features, Bioinformatics, 30, 923, 10.1093/bioinformatics/btt656

MI Love, 2014, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, 15, 550, 10.1186/s13059-014-0550-8

R Herwig, 2016, Analyzing and interpreting genome data at the network level with ConsensusPathDB, Nat Protoc, 11, 1889, 10.1038/nprot.2016.117