Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38772371

ABSTRACT

Peripheral sensory neurons widely innervate various tissues to continuously monitor and respond to environmental stimuli. Whether peripheral sensory neurons innervate the spleen and modulate splenic immune response remains poorly defined. Here, we demonstrate that nociceptive sensory nerve fibers extensively innervate the spleen along blood vessels and reach B cell zones. The spleen-innervating nociceptors predominantly originate from left T8-T13 dorsal root ganglia (DRGs), promoting the splenic germinal center (GC) response and humoral immunity. Nociceptors can be activated by antigen-induced accumulation of splenic prostaglandin E2 (PGE2) and then release calcitonin gene-related peptide (CGRP), which further promotes the splenic GC response at the early stage. Mechanistically, CGRP directly acts on B cells through its receptor CALCRL-RAMP1 via the cyclic AMP (cAMP) signaling pathway. Activating nociceptors by ingesting capsaicin enhances the splenic GC response and anti-influenza immunity. Collectively, our study establishes a specific DRG-spleen sensory neural connection that promotes humoral immunity, suggesting a promising approach for improving host defense by targeting the nociceptive nervous system.

2.
Science ; 380(6648): 972-979, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37262147

ABSTRACT

The suprachiasmatic nucleus (SCN) drives circadian clock coherence through intercellular coupling, which is resistant to environmental perturbations. We report that primary cilia are required for intercellular coupling among SCN neurons to maintain the robustness of the internal clock in mice. Cilia in neuromedin S-producing (NMS) neurons exhibit pronounced circadian rhythmicity in abundance and length. Genetic ablation of ciliogenesis in NMS neurons enabled a rapid phase shift of the internal clock under jet-lag conditions. The circadian rhythms of individual neurons in cilia-deficient SCN slices lost their coherence after external perturbations. Rhythmic cilia changes drive oscillations of Sonic Hedgehog (Shh) signaling and clock gene expression. Inactivation of Shh signaling in NMS neurons phenocopied the effects of cilia ablation. Thus, cilia-Shh signaling in the SCN aids intercellular coupling.


Subject(s)
Cilia , Circadian Clocks , Circadian Rhythm , Hedgehog Proteins , Suprachiasmatic Nucleus Neurons , Animals , Mice , Cilia/metabolism , Cilia/physiology , Circadian Clocks/genetics , Circadian Rhythm/physiology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Suprachiasmatic Nucleus Neurons/physiology , Signal Transduction , Gene Expression Regulation , Mice, Transgenic
3.
J Cell Biol ; 221(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34813648

ABSTRACT

Primary cilia transduce diverse signals in embryonic development and adult tissues. Defective ciliogenesis results in a series of human disorders collectively known as ciliopathies. The CP110-CEP97 complex removal from the mother centriole is an early critical step for ciliogenesis, but the underlying mechanism for this step remains largely obscure. Here, we reveal that the linear ubiquitin chain assembly complex (LUBAC) plays an essential role in ciliogenesis by targeting the CP110-CEP97 complex. LUBAC specifically generates linear ubiquitin chains on CP110, which is required for CP110 removal from the mother centriole in ciliogenesis. We further identify that a pre-mRNA splicing factor, PRPF8, at the distal end of the mother centriole acts as the receptor of the linear ubiquitin chains to facilitate CP110 removal at the initial stage of ciliogenesis. Thus, our study reveals a direct mechanism of regulating CP110 removal in ciliogenesis and implicates the E3 ligase LUBAC as a potential therapy target of cilia-associated diseases, including ciliopathies and cancers.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Cilia/metabolism , Microtubule-Associated Proteins/metabolism , Organogenesis , Phosphoproteins/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Humans , Mice , Multiprotein Complexes , RNA-Binding Proteins/metabolism , Substrate Specificity , Ubiquitination , Zebrafish
4.
Nat Commun ; 12(1): 662, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510165

ABSTRACT

Dynamic assembly and disassembly of primary cilia controls embryonic development and tissue homeostasis. Dysregulation of ciliogenesis causes human developmental diseases termed ciliopathies. Cell-intrinsic regulatory mechanisms of cilia disassembly have been well-studied. The extracellular cues controlling cilia disassembly remain elusive, however. Here, we show that lysophosphatidic acid (LPA), a multifunctional bioactive phospholipid, acts as a physiological extracellular factor to initiate cilia disassembly and promote neurogenesis. Through systematic analysis of serum components, we identify a small molecular-LPA as the major driver of cilia disassembly. Genetic inactivation and pharmacological inhibition of LPA receptor 1 (LPAR1) abrogate cilia disassembly triggered by serum. The LPA-LPAR-G-protein pathway promotes the transcription and phosphorylation of cilia disassembly factors-Aurora A, through activating the transcription coactivators YAP/TAZ and calcium/CaM pathway, respectively. Deletion of Lpar1 in mice causes abnormally elongated cilia and decreased proliferation in neural progenitor cells, thereby resulting in defective neurogenesis. Collectively, our findings establish LPA as a physiological initiator of cilia disassembly and suggest targeting the metabolism of LPA and the LPA pathway as potential therapies for diseases with dysfunctional ciliogenesis.


Subject(s)
Cilia/drug effects , Lysophospholipids/pharmacology , Neurogenesis/drug effects , Retinal Pigment Epithelium/drug effects , Signal Transduction , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cilia/genetics , Cilia/metabolism , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Lysophospholipids/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/genetics , Protein Binding , RNA Interference , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism
5.
J Cell Biol ; 220(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33475699

ABSTRACT

Primary cilia protrude from the cell surface and have diverse roles during development and disease, which depends on the precise timing and control of cilia assembly and disassembly. Inactivation of assembly often causes cilia defects and underlies ciliopathy, while diseases caused by dysfunction in disassembly remain largely unknown. Here, we demonstrate that CEP55 functions as a cilia disassembly regulator to participate in ciliopathy. Cep55-/- mice display clinical manifestations of Meckel-Gruber syndrome, including perinatal death, polycystic kidneys, and abnormalities in the CNS. Interestingly, Cep55-/- mice exhibit an abnormal elongation of cilia on these tissues. Mechanistically, CEP55 promotes cilia disassembly by interacting with and stabilizing Aurora A kinase, which is achieved through facilitating the chaperonin CCT complex to Aurora A. In addition, CEP55 mutation in Meckel-Gruber syndrome causes the failure of cilia disassembly. Thus, our study establishes a cilia disassembly role for CEP55 in vivo, coupling defects in cilia disassembly to ciliopathy and further suggesting that proper cilia dynamics are critical for mammalian development.


Subject(s)
Aurora Kinase A/metabolism , Cell Cycle Proteins/metabolism , Cilia/metabolism , Animals , Cell Cycle Checkpoints , Cell Cycle Proteins/deficiency , Cells, Cultured , Centrosome/metabolism , Centrosome/ultrastructure , Chaperonin Containing TCP-1/metabolism , Cilia/ultrastructure , Ciliary Motility Disorders/pathology , Encephalocele/pathology , Enzyme Stability , Gene Targeting , HEK293 Cells , Humans , Mice , Mitosis , Phenotype , Polycystic Kidney Diseases/pathology , Protein Binding , Retinitis Pigmentosa/pathology , Smoothened Receptor/metabolism
6.
J Cell Biol ; 218(12): 4030-4041, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31619485

ABSTRACT

The primary cilium is a sensory organelle that protrudes from the cell surface. Primary cilia undergo dynamic transitions between assembly and disassembly to exert their function in cell signaling. In this study, we identify the small GTPase Rab7 as a novel regulator of cilia disassembly. Depletion of Rab7 potently induced spontaneous ciliogenesis in proliferating cells and promoted cilia elongation during quiescence. Moreover, Rab7 performs an essential role in cilia disassembly; knockdown of Rab7 blocked serum-induced ciliary resorption, and active Rab7 was required for this process. Further, we demonstrate that Rab7 depletion significantly suppresses cilia tip excision, referred to as cilia ectocytosis, which has been identified as required for cilia disassembly. Mechanically, the failure of F-actin polymerization at the site of excision of cilia tips caused suppression of cilia ectocytosis on Rab7 depletion. Overall, our results suggest a novel function for Rab7 in regulating cilia ectocytosis and cilia disassembly via control of intraciliary F-actin polymerization.


Subject(s)
Actin Cytoskeleton/metabolism , Cilia/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism , Actins/metabolism , Cell Division , Cell Line , Cell Proliferation , GTP Phosphohydrolases/metabolism , HEK293 Cells , Humans , Maltose-Binding Proteins/metabolism , Polymers/metabolism , RNA, Small Interfering/metabolism , rab7 GTP-Binding Proteins
7.
Cell ; 178(1): 176-189.e15, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31155231

ABSTRACT

RLR-mediated type I IFN production plays a pivotal role in elevating host immunity for viral clearance and cancer immune surveillance. Here, we report that glycolysis, which is inactivated during RLR activation, serves as a barrier to impede type I IFN production upon RLR activation. RLR-triggered MAVS-RIG-I recognition hijacks hexokinase binding to MAVS, leading to the impairment of hexokinase mitochondria localization and activation. Lactate serves as a key metabolite responsible for glycolysis-mediated RLR signaling inhibition by directly binding to MAVS transmembrane (TM) domain and preventing MAVS aggregation. Notably, lactate restoration reverses increased IFN production caused by lactate deficiency. Using pharmacological and genetic approaches, we show that lactate reduction by lactate dehydrogenase A (LDHA) inactivation heightens type I IFN production to protect mice from viral infection. Our study establishes a critical role of glycolysis-derived lactate in limiting RLR signaling and identifies MAVS as a direct sensor of lactate, which functions to connect energy metabolism and innate immunity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism , Lactic Acid/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Animals , Female , Glycolysis , HEK293 Cells , Humans , Interferon-beta/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RAW 264.7 Cells , Receptors, Immunologic , Signal Transduction/drug effects , Transfection
8.
Nat Commun ; 10(1): 1839, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015398

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) are capable of producing all mature blood lineages, as well as maintaining the self-renewal ability throughout life. The hairy-like organelle, cilium, is present in most types of vertebrate cells, and plays important roles in various biological processes. However, it is unclear whether and how cilia regulate HSPC development in vertebrates. Here, we show that cilia-specific genes, involved in primary cilia formation and function, are required for HSPC development, especially in hemogenic endothelium (HE) specification in zebrafish embryos. Blocking primary cilia formation or function by genetic or chemical manipulations impairs HSPC development. Mechanistically, we uncover that primary cilia in endothelial cells transduce Notch signal to the earliest HE for proper HSPC specification during embryogenesis. Altogether, our findings reveal a pivotal role of endothelial primary cilia in HSPC development, and may shed lights into in vitro directed differentiation of HSPCs.


Subject(s)
Cilia/metabolism , Hematopoietic Stem Cells/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cilia/genetics , Embryo, Nonmammalian , Embryonic Development/physiology , Hemangioblasts/cytology , Hemangioblasts/metabolism , Hematopoiesis/physiology , Models, Animal , Zebrafish/physiology
9.
Nat Commun ; 10(1): 273, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30655516

ABSTRACT

Faithful chromosome segregation requires proper chromosome congression at prometaphase and dynamic maintenance of the aligned chromosomes at metaphase. Chromosome missegregation can result in aneuploidy, birth defects and cancer. The kinetochore-bound KMN network and the kinesin motor CENP-E are critical for kinetochore-microtubule attachment and chromosome stability. The linear ubiquitin chain assembly complex (LUBAC) attaches linear ubiquitin chains to substrates, with well-established roles in immune response. Here, we identify LUBAC as a key player of chromosome alignment during mitosis. LUBAC catalyzes linear ubiquitination of the kinetochore motor CENP-E, which is specifically required for the localization of CENP-E at attached kinetochores, but not unattached ones. KNL1 acts as a receptor of linear ubiquitin chains to anchor CENP-E at attached kinetochores in prometaphase and metaphase. Thus, linear ubiquitination promotes chromosome congression and dynamic chromosome alignment by coupling the dynamic kinetochore microtubule receptor CENP-E to the static one, the KMN network.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Carrier Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , Fibroblasts , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Mitosis , Primary Cell Culture , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
10.
Nat Commun ; 9(1): 5277, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30538248

ABSTRACT

Defective ciliogenesis causes human developmental diseases termed ciliopathies. Microtubule (MT) asters originating from centrosomes in mitosis ensure the fidelity of cell division by positioning the spindle apparatus. However, the function of microtubule asters in interphase remains largely unknown. Here, we reveal an essential role of MT asters in transition zone (TZ) assembly during ciliogenesis. We demonstrate that the centrosome protein FSD1, whose biological function is largely unknown, anchors MT asters to interphase centrosomes by binding to microtubules. FSD1 knockdown causes defective ciliogenesis and affects embryonic development in vertebrates. We further show that disruption of MT aster anchorage by depleting FSD1 or other known anchoring proteins delocalizes the TZ assembly factor Cep290 from centriolar satellites, and causes TZ assembly defects. Thus, our study establishes FSD1 as a MT aster anchorage protein and reveals an important function of MT asters anchored by FSD1 in TZ assembly during ciliogenesis.


Subject(s)
Axoneme/metabolism , Cilia/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Animals , Axoneme/genetics , Centrosome/metabolism , Cilia/genetics , Humans , Mitosis , Nerve Tissue Proteins/genetics , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
11.
Oncotarget ; 8(2): 2197-2208, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27517492

ABSTRACT

Excessive accumulation of DNA damage will generate chromosome stress, leading to various chromosome abnormalities such as chromatin bridge and result in genomic instability. Orchestra procession and regulation of DNA damage repair are vital for keeping genome stability. Despite of the key role of HDAC1/2 in double strand break (DSB) repair, the regulation for their mode of action is less well understood. In this study, we found that deubiquitination enzymes USP19 physically interacts with HDAC1/2 and specifically regulate their K63-linked ubiquitination, which might be crucial for regulation of HDAC1/2 activity in DNA damage repair. Notably, we found that USP19 trans-locate into nucleus upon IR irradiation and is indispensable for normally DNA damage response. In addition, we showed that USP19 play critical role in preventing anaphase bridge formation through regulating DNA damage repair process. Furthermore, the expression level of USP19 is commonly lower or deleted in several types of tumor. These results indicated that USP19 is a key factor in modulating DNA damage repair by targeting HDAC1/2 K63-linked ubiquitination, cells with deletion or decreased expression of USP19 might cause genome instability and even contribute to tumorigenesis.


Subject(s)
DNA Repair/genetics , Endopeptidases/physiology , Genomic Instability/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Ubiquitination , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , DNA Damage/genetics , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Protein Processing, Post-Translational
12.
FEBS Lett ; 589(19 Pt B): 2850-8, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26299341

ABSTRACT

Most of NF-κB (nuclear factor kappa B) signaling molecules have various types of post-translational modifications. In this study, we focused on ubiquitination and designed a siRNA library including most ubiquitin-binding domains. With this library, we identified several candidate regulators of canonical NF-κB pathway, including RNF4. Overexpression of RNF4 impaired NF-κB activation in a dose-dependent manner, whereas RNF4 knockdown potentiated NF-κB activation. We showed that RNF4 interacts with the TAK1-TAB2-TAB3 complex, but not TAB1. Further, we found that RNF4 specifically down-regulated TAB2 through a lysosomal pathway, and knockdown of RNF4 impaired endogenous TAB2 degradation. Therefore, our findings will provide new insights into the negative regulation of NF-κB signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Down-Regulation , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Gene Knockdown Techniques , Humans , Interleukin-1beta/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Protein Structure, Tertiary , Proteolysis/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/genetics , Ubiquitin/metabolism
13.
J Biol Chem ; 290(16): 10395-405, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25681446

ABSTRACT

Excessive nuclear factor κB (NF-κB) activation should be precisely controlled as it contributes to multiple immune and inflammatory diseases. However, the negative regulatory mechanisms of NF-κB activation still need to be elucidated. Various types of polyubiquitin chains have proved to be involved in the process of NF-κB activation. Many negative regulators linked to ubiquitination, such as A20 and CYLD, inhibit IκB kinase activation in the NF-κB signaling pathway. To find new NF-κB signaling regulators linked to ubiquitination, we used a small scale siRNA library against 51 ubiquitin-associated domain-containing proteins and screened out UBXN1, which contained both ubiquitin-associated and ubiquitin regulatory X (UBX) domains as a negative regulator of TNFα-triggered NF-κB activation. Overexpression of UBXN1 inhibited TNFα-triggered NF-κB activation, although knockdown of UBXN1 had the opposite effect. UBX domain-containing proteins usually act as valosin-containing protein (VCP)/p97 cofactors. However, knockdown of VCP/p97 barely affected UBXN1-mediated NF-κB inhibition. At the same time, we found that UBXN1 interacted with cellular inhibitors of apoptosis proteins (cIAPs), E3 ubiquitin ligases of RIP1 in the TNFα receptor complex. UBXN1 competitively bound to cIAP1, blocked cIAP1 recruitment to TNFR1, and sequentially inhibited RIP1 polyubiquitination in response to TNFα. Therefore, our findings demonstrate that UBXN1 is an important negative regulator of the TNFα-triggered NF-κB signaling pathway by mediating cIAP recruitment independent of VCP/p97.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation , NF-kappa B/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Proteolysis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Small Molecule Libraries , Tumor Necrosis Factor-alpha/pharmacology , Valosin Containing Protein
14.
Opt Lett ; 39(19): 5705-8, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25360964

ABSTRACT

For the first time, transparent La0.2Y1.8O3 nanostructured polycrystalline scintillators were fabricated by sintering nanoparticle powders at high temperatures and their scintillation properties are reported. La0.2Y1.8O3 is a host material that has never been investigated as scintillators for radiation detection. Our observations found that La0.2Y1.8O3 has an intense scintillation luminescence, a detection efficiency higher than that of YAG:Ce and a comparable energy resolution to NaI and CsI scintillators. In addition, La0.2Y1.8O3 is stable and has luminescence decay lifetime in the picosecond range which is favorable for radiation detection. The luminescence of La0.2Y1.8O3 has a large Stokes-shift and a large emission bandwidth, and the luminescence is highly temperature dependent. Different from most doped scintillators, the luminescence of La0.2Y1.8O3 is most likely from the self-trapped excitons. The discovery of La0.2Y1.8O3 scintillators opens a new door for the research of new materials for radiation detection.

15.
J Nanosci Nanotechnol ; 14(5): 3965-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24734674

ABSTRACT

Nanocrystalline yttria-stabilized zirconia (ZrO2-8 mol%Y2O3, 8YSZ) was synthesized by a homogeneous precipitation process using urea as the precipitation agent. Zirconia (ZrO2) transparent ceramics samples have been successfully fabricated by a microwave sintering process at low temperature. The technologies of low-temperature microwave sintering and the relationships of the microstructures and properties of the specified samples have been investigated in detail. We have found out that the low-temperature microwave sintering has its obvious advantages over the other methods in manufacturing zirconia transparent ceramics.

SELECTION OF CITATIONS
SEARCH DETAIL
...