Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cancer Immunol Immunother ; 73(8): 143, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38832955

ABSTRACT

This study investigates the role of USP47, a deubiquitinating enzyme, in the tumor microenvironment and its impact on antitumor immune responses. Analysis of TCGA database revealed distinct expression patterns of USP47 in various tumor tissues and normal tissues. Prostate adenocarcinoma showed significant downregulation of USP47 compared to normal tissue. Correlation analysis demonstrated a positive association between USP47 expression levels and infiltrating CD8+ T cells, neutrophils, and macrophages, while showing a negative correlation with NKT cells. Furthermore, using Usp47 knockout mice, we observed a slower tumor growth rate and reduced tumor burden. The absence of USP47 led to increased infiltration of immune cells, including neutrophils, macrophages, NK cells, NKT cells, and T cells. Additionally, USP47 deficiency resulted in enhanced activation of cytotoxic T lymphocytes (CTLs) and altered T cell subsets within the tumor microenvironment. These findings suggest that USP47 plays a critical role in modulating the tumor microenvironment and promoting antitumor immune responses, highlighting its potential as a therapeutic target in prostate cancer.


Subject(s)
Lymphocytes, Tumor-Infiltrating , Prostatic Neoplasms , Animals , Humans , Male , Mice , Cell Line, Tumor , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Mice, Knockout , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tumor Microenvironment
2.
Proc Natl Acad Sci U S A ; 110(16): 6459-64, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23553835

ABSTRACT

Retinoic acid inducible gene I (RIG-I) senses viral RNAs and triggers innate antiviral responses through induction of type I IFNs and inflammatory cytokines. However, whether RIG-I interacts with host cellular RNA remains undetermined. Here we report that Rig-I interacts with multiple cellular mRNAs, especially Nf-κb1. Rig-I is required for NF-κB activity via regulating Nf-κb1 expression at posttranscriptional levels. It interacts with the multiple binding sites within 3'-UTR of Nf-κb1 mRNA. Further analyses reveal that three distinct tandem motifs enriched in the 3'-UTR fragments can be recognized by Rig-I. The 3'-UTR binding with Rig-I plays a critical role in normal translation of Nf-κb1 by recruiting the ribosomal proteins [ribosomal protein L13 (Rpl13) and Rpl8] and rRNAs (18S and 28S). Down-regulation of Rig-I or Rpl13 significantly reduces Nf-κb1 and 3'-UTR-mediated luciferase expression levels. These findings indicate that Rig-I functions as a positive regulator for NF-κB signaling and is involved in multiple biological processes in addition to host antivirus immunity.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Expression Regulation/physiology , NF-kappa B/metabolism , RNA, Messenger/metabolism , 3' Untranslated Regions/genetics , Animals , Blotting, Northern , Blotting, Western , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Fluorescent Antibody Technique , Immunoprecipitation , Luciferases , Mice , Mice, Knockout , Microarray Analysis , Molecular Dynamics Simulation , NF-kappa B/genetics , RNA Interference , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/metabolism
3.
Am J Hum Genet ; 91(6): 1088-94, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23141294

ABSTRACT

Charcot-Marie-Tooth (CMT) disease represents a clinically and genetically heterogeneous group of inherited neuropathies. Here, we report a five-generation family of eight affected individuals with CMT disease type 2, CMT2. Genome-wide linkage analysis showed that the disease phenotype is closely linked to chromosomal region 10p13-14, which spans 5.41 Mb between D10S585 and D10S1477. DNA-sequencing analysis revealed a nonsense mutation, c.1455T>G (p.Tyr485(∗)), in exon 8 of dehydrogenase E1 and transketolase domain-containing 1 (DHTKD1) in all eight affected individuals, but not in other unaffected individuals in this family or in 250 unrelated normal persons. DHTKD1 mRNA expression levels in peripheral blood of affected persons were observed to be half of those in unaffected individuals. In vitro studies have shown that, compared to wild-type mRNA and DHTKD1, mutant mRNA and truncated DHTKD1 are significantly decreased by rapid mRNA decay in transfected cells. Inhibition of nonsense-mediated mRNA decay by UPF1 silencing effectively rescued the decreased levels of mutant mRNA and protein. More importantly, DHTKD1 silencing was found to lead to impaired energy production, evidenced by decreased ATP, total NAD(+) and NADH, and NADH levels. In conclusion, our data demonstrate that the heterozygous nonsense mutation in DHTKD1 is one of CMT2-causative genetic alterations, implicating an important role for DHTKD1 in mitochondrial energy production and neurological development.


Subject(s)
Asian People/genetics , Charcot-Marie-Tooth Disease/genetics , Codon, Nonsense , Ketone Oxidoreductases/genetics , Amino Acid Sequence , Base Sequence , Charcot-Marie-Tooth Disease/diagnosis , Charcot-Marie-Tooth Disease/metabolism , China , Exons , Female , Gene Order , Humans , Ketoglutarate Dehydrogenase Complex , Male , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Nonsense Mediated mRNA Decay , Pedigree
4.
Proc Natl Acad Sci U S A ; 108(29): 12001-6, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730163

ABSTRACT

There are currently two distinct models proposed to explain why both MDM2 and MDMX are required in p53 control, with a key difference centered on whether these two p53 inhibitors work together or independently. To test these two competing models, we generated knockin mice expressing a point mutation MDMX mutant (C462A) that is defective in MDM2 binding. This approach allowed a targeted disassociation of the MDM2/MDMX heterocomplex without affecting the ability of MDMX to bind to p53, and while leaving the MDM2 protein itself completely untouched. Significantly, Mdmx(C462A/C462A) homozygous mice died at approximately day 9.5 of embryonic development, as the result of a combination of apoptosis and decreased cell proliferation, as shown by TUNEL and BrdU incorporation assays, respectively. Interestingly, even though the MDMX mutant protein abundance was found slightly elevated in the Mdmx(C462A/C462A) homozygous embryos, both the abundance and activity of p53 were markedly increased. A p53-dependent death was demonstrated by the finding that concomitant deletion of p53 completely rescued the embryonic lethality in Mdmx(C462A/C462A) homozygous mice. Our data demonstrate that MDM2 and MDMX function as an integral complex in p53 control, providing insights into the nonredundant nature of the function of MDM2 and MDMX.


Subject(s)
Gene Expression Regulation/genetics , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Apoptosis/genetics , Blotting, Western , Bromodeoxyuridine , Gene Knock-In Techniques , Genotype , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Mutation/genetics , Polymerase Chain Reaction , Proto-Oncogene Proteins c-mdm2/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Proc Natl Acad Sci U S A ; 108(5): 1897-902, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21224412

ABSTRACT

RA-inducible gene I (RIG-I/DDX58) has been shown to activate IFN-ß promoter stimulator 1 (IPS-1) on recognizing cytoplasmic viral RNAs. It is unclear how RIG-I functions within the IFN and/or RA signaling process in acute myeloid leukemia (AML) cells, however, where obvious RIG-I induction is observed. Here, we show that the RIG-I induction functionally contributes to IFN-α plus RA-triggered growth inhibition of AML cells. Interestingly, although RIG-I induction itself is under the regulation of STAT1, a major IFN intracellular signal mediator, under circumstances in which it does not stimulate IPS-1, it conversely augments STAT1 activation to induce IFN-stimulatory gene expression and inhibit leukemia cell proliferation. Thus, our results unveil a previously undescribed RIG-I activity in regulating the cellular proliferation of leukemia cells via STAT1, which is independent of its classic role of sensing viral invasion to trigger type I IFN transcription.


Subject(s)
Cell Proliferation , DEAD-box RNA Helicases/genetics , Leukemia/pathology , STAT1 Transcription Factor/metabolism , DEAD Box Protein 58 , Gene Expression Regulation/drug effects , Humans , Receptors, Immunologic , Tretinoin/pharmacology , U937 Cells
6.
BMC Neurosci ; 14: 76, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902361

ABSTRACT

BACKGROUND: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces Parkinson's disease (PD)-like neurodegeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) via its oxidized product, 1-methyl-4-phenylpyridinium (MPP+), which is transported by the dopamine (DA) transporter into DA nerve terminals. DA receptor subtype 3 (D3 receptor) participates in neurotransmitter transport, gene regulation in the DA system, physiological accommodation via G protein-coupled superfamily receptors and other physiological processes in the nervous system. This study investigated the possible correlation between D3 receptors and MPTP-induced neurotoxicity. A series of behavioral experiments and histological analyses were conducted in D3 receptor-deficient mice, using an MPTP-induced model of PD. RESULTS: After the fourth MPTP injection, wild-type animals that received 15 mg/kg per day displayed significant neurotoxin-related bradykinesia. D3 receptor-deficient mice displayed attenuated MPTP-induced locomotor activity changes. Consistent with the behavioral observations, further neurohistological assessment showed that MPTP-induced neuronal damage in the SNpc was reduced in D3 receptor-deficient mice. CONCLUSIONS: Our study indicates that the D3 receptor might be an essential molecule in MPTP-induced PD and provides a new molecular mechanism for MPTP neurotoxicity.


Subject(s)
MPTP Poisoning/metabolism , MPTP Poisoning/physiopathology , Receptors, Dopamine D3/physiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Analysis of Variance , Animals , Disease Models, Animal , Drug Administration Schedule , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , MPTP Poisoning/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Psychomotor Performance/drug effects , Receptors, Dopamine D3/deficiency , Time Factors , Tyrosine 3-Monooxygenase/metabolism
7.
Proc Natl Acad Sci U S A ; 107(7): 2956-61, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133625

ABSTRACT

HYPB is a human histone H3 lysine 36 (H3K36)-specific methyltransferase and acts as the ortholog of yeast Set2. This study explored the physiological function of mammalian HYPB using knockout mice. Homozygous disruption of Hypb impaired H3K36 trimethylation but not mono- or dimethylation, and resulted in embryonic lethality at E10.5-E11.5. Severe vascular defects were observed in the Hypb(-/-) embryo, yolk sac, and placenta. The abnormally dilated capillaries in mutant embryos and yolk sacs could not be remodeled into large blood vessels or intricate networks, and the aberrantly rounded mesodermal cells exhibited weakened interaction with endothelial cells. The embryonic vessels failed to invade the labyrinthine layer of placenta, which impaired the embryonic-maternal vascular connection. These defects could not be rescued by wild-type tetraploid blastocysts, excluding the possibility that they were caused by the extraembryonic tissues. Consistent with these phenotypes, gene expression profiling in wild-type and Hypb(-/-) yolk sacs revealed that the Hypb disruption altered the expression of some genes involved in vascular remodeling. At the cellular level, Hypb(-/-) embryonic stem cell-derived embryonic bodies, as well as in vitro-cultured human endothelial cells with siRNA-mediated suppression of HYPB, showed obvious defects in cell migration and invasion during vessel formation, suggesting an intrinsic role of Hypb in vascular development. Taken together, these results indicate that Hypb is required for embryonic vascular remodeling and provide a tool to study the function of H3K36 methylation in vasculogenesis/angiogenesis.


Subject(s)
Embryo, Mammalian/physiology , Gene Expression Regulation, Developmental/physiology , Histone-Lysine N-Methyltransferase/metabolism , Neovascularization, Physiologic/physiology , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Gene Expression Profiling , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , RNA Interference
8.
Life Sci ; 334: 122241, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37944639

ABSTRACT

AIMS: TAFA2, a cytokine specifically expressed in the central nervous system, plays a vital role in neuronal cell survival. TAFA2 deficiency has been correlated to various neurological disorders in mice and humans. However, the underlying mechanism remains elusive, especially its membrane-binding receptor through which TAFA2 functions. This study aimed to identify the specific binding receptor responsible for the anti-apoptotic effects of TAFA2. MAIN METHOD: Co-immunoprecipitation (Co-IP) and quantitative mass spectrometry-based proteomic analysis were employed to identify potential TAFA2 binding proteins in V5 knockin mouse brain lysates. Subsequent validation involved in vitro and in vivo Co-IP and pull-down using specific antibodies. The functional analysis included evaluating the effects of ADGRL1 knockout, overexpression, and Lectin-like domain (Lec) deletion mutant on TAFA2's anti-apoptotic activity and analyzing the intracellular signaling pathways mediated by TAFA2 through ADGRL1. KEY FINDINGS: Our study identified ADGRL1 as a potential receptor for TAFA2, which directly binds to TAFA2 through its lectin-like domain. Overexpression ADGRL1, but not ADGRL1ΔLec, induced apoptosis, which could be effectively suppressed by recombinant TAFA2 (rTAFA2). In ADGRL1-/- cells or re-introducing with ADGRL1ΔLec, responses to rTAFA2 in suppressing cell apoptosis were compromised. Increased cAMP, p-PKA, p-CREB, and BCL2 levels were also observed in response to rTAFA2 treatment, with these responses attenuated in ADGRL1-/- or ADGRL1ΔLec-expressing cells. SIGNIFICANCE: Our results demonstrated that TAFA2 directly binds to the lectin-like domain of ADGRL1, activating cAMP/PKA/CREB/BCL2 signaling pathway, which is crucial in preventing cell death. These results implicate TAFA2 and its receptor ADGRL1 as potential therapeutic targets for neurological disorders.


Subject(s)
Nervous System Diseases , Proteomics , Animals , Humans , Mice , Apoptosis , Cell Survival , Cyclic AMP Response Element-Binding Protein/metabolism , Lectins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
9.
Am J Hum Genet ; 85(1): 53-63, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19589401

ABSTRACT

Fibroblast growth factors (FGFs) play diverse roles in several developmental processes. Mutations leading to deregulated FGF signaling can cause human skeletal dysplasias and cancer.(1,2) Here we report a missense mutation (Ser99Asp) in exon 2 of FGF9 in 12 patients with multiple synostoses syndrome (SYNS) in a large Chinese family. In vitro studies demonstrate that FGF9(S99N) is expressed and secreted as efficiently as wild-type FGF9 in transfected cells. However, FGF9(S99N) induces compromised chondrocyte proliferation and differentiation, which is accompanied by enhanced osteogenic differentiation and matrix mineralization of bone marrow-derived mesenchymal stem cells (BMSCs). Biochemical analysis reveals that S99N mutation in FGF9 leads to significantly impaired FGF signaling, as evidenced by diminished activity of Erk1/2 pathway and decreased beta-catenin and c-Myc expression when compared with wild-type FGF9. Importantly, the binding of FGF9(S99N) to its receptor is severely impaired although the dimerization ability of mutant FGF9 itself or with wild-type FGF9 is not detectably affected, providing a basis for the defective FGFR signaling. Collectively, our data demonstrate a previously uncharacterized mutation in FGF9 as one of the causes of SYNS, implicating an important role of FGF9 in normal joint development.


Subject(s)
Exons , Fibroblast Growth Factor 9/genetics , Mutation, Missense , Synostosis/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , Child , Child, Preschool , DNA Mutational Analysis , Female , Fibroblast Growth Factor 9/chemistry , Humans , Male , Middle Aged , Molecular Sequence Data , Pedigree , Signal Transduction
10.
Neuroscience ; 503: 131-145, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36115515

ABSTRACT

Adhesion G protein-coupled receptor A1 (ADGRA1) belongs to the G protein-coupled receptor (GPCR) family, and its physiological function remains largely unknown. We found that Adgra1 is highly and exclusively expressed in the brain, suggesting that Adgra1 may be involved in the regulation of neurological behaviors including anxiety, depression, learning and memory. To this end, we comprehensively analyzed the potential role of ADGRA1 in the neurobehaviors of mice by comparing Adgra1-/- and their wild-type (wt) littermates. We found that Adgra1-/- male but not female mice exhibited elevated anxiety levels in the open field, elevated plus maze, and light-dark box tests, with normal depression levels in the tail-suspension and forced-swim tests, and comparable learning and memory abilities in the Morris water maze, Y maze, fear condition, and step-down avoidance tests. Further studies showed that ADGRA1 deficiency resulted in higher dendritic branching complexity and spine density as evidenced by elevated expression levels of SYN and PSD95 in amygdalae of male mice. Finally, we found that PI3K/AKT/GSK-3ß and MEK/ERK in amygdalae of Adgra1-deficient male mice were aberrantly activated when compared to wt male mice. Together, our findings reveal an important suppressive role of ADGRA1 in anxiety control and synaptic function by regulating the PI3K/AKT/GSK-3ß and MEK/ERK pathways in amygdalae of male mice, implicating a potential, therapeutic application in novel anti-anxiety drug development.


Subject(s)
Anti-Anxiety Agents , Phosphatidylinositol 3-Kinases , Animals , Male , Mice , Dendrites/metabolism , Disks Large Homolog 4 Protein/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/metabolism
11.
Biology (Basel) ; 11(11)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36421382

ABSTRACT

Serine-threonine kinase 10 (STK10) is a member of the STE20/p21-activated kinase (PAK) family and is predominantly expressed in immune organs. Our previous reports suggested that STK10 participates in the growth and metastasis of prostate cancer via in vitro and in vivo data. However, the correlation between STK10 and the tumor microenvironment (TME) remains unclear. In this study, we assessed the relationship between STK10 and the immune cells in the tumor microenvironment of prostate cancer through bioinformatic analysis, and investigated the role of Stk10 in tumor growth using an Stk10 knockout mouse model. The results showed that STK10 is significantly associated with the tumor-infiltrating immune cells including lymphocytes, neutrophils, macrophages and dendritic cells. The target deletion of host Stk10 results in increased tumor growth, due to decreased activated/effector cytotoxic T lymphocytes (CTLs) and increased vessel density in the TME. In conclusion, we demonstrate that host Stk10 is involved in the host anti-tumor response by modulating the activated tumor-infiltrated CTLs and angiogenesis.

12.
Hum Cell ; 35(4): 1071-1083, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35416622

ABSTRACT

Studies have indicated that RIG-I may act as a tumor suppressor and participate in the tumorigenesis of some malignant diseases. However, RIG-I induces distinct cellular responses via different downstream signaling pathways depending on the cell type. To investigate the biological function and underlying molecular mechanism of RIG-I in the tumorigenesis of melanoma, we constructed RIG-I knockout, RIG-I-overexpressing B16-F10 and RIG-I knockdown A375 melanoma cell lines, and analyzed the RIG-I-mediated change in the biological behavior of tumor cells in spontaneous and poly (I:C)-induced RIG-I activation. Cell proliferation, cell cycling, apoptosis and migration were detected by CCK-8 assay, BrdU incorporation assay, Annexin V-PI staining assay and Transwell assay, respectively. In vivo tumorigenicity was evaluated by tumor xenograft growth in nude mice and subsequently by Ki67 staining and TUNEL assays. Furthermore, Western blotting was utilized to explore the underlying mechanism of RIG-I in melanoma cells. Our data showed that RIG-I promotes apoptosis and inhibits proliferation by G1 phase cell cycle arrest in the melanoma cell lines. Mechanistically, RIG-I induced the phosphorylation of p38 MAPK and MAPK kinases MKK3 and MKK4. In conclusion, the current study demonstrated that RIG-I suppressed the development of melanoma by regulating the activity of the MKK/p38 MAPK signaling pathway, which is relevant to research on novel therapeutic targets for this malignant disease.


Subject(s)
DEAD Box Protein 58 , Melanoma , Mitogen-Activated Protein Kinase Kinases , Receptors, Immunologic , Skin Neoplasms , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Humans , Melanoma/genetics , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Receptors, Immunologic/genetics , Signal Transduction/genetics , Skin Neoplasms/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Proc Natl Acad Sci U S A ; 105(30): 10553-8, 2008 Jul 29.
Article in English | MEDLINE | ID: mdl-18650396

ABSTRACT

RIG-I has been implicated in innate immunity by sensing intracellular viral RNAs and inducing type I IFN production. However, we have found a significant RIG-I induction in a biological setting without active viral infection-namely, during RA-induced terminal granulocytic differentiation of acute myeloid leukemias. Here, we present evidence that a significant Rig-I induction also occurs during normal myelopoiesis and that the disruption of the Rig-I gene in mice leads to the development of a progressive myeloproliferative disorder. The initiation of progressive myeloproliferative disorder is mainly due to an intrinsic defect of Rig-I(-/-) myeloid cells, which are characterized by a reduced expression of IFN consensus sequence binding protein, a major regulator of myeloid differentiation. Thus, our study reveals a critical regulatory role of Rig-I in modulating the generation and differentiation of granulocytes.


Subject(s)
DEAD-box RNA Helicases/physiology , Gene Expression Regulation , Granulocytes/cytology , Receptors, Retinoic Acid/physiology , Animals , Cell Differentiation , Cell Proliferation , DEAD Box Protein 58 , Exons , Granulocytes/metabolism , Hematopoietic Stem Cells/cytology , Humans , Immunity, Innate , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Cells/cytology , Myeloproliferative Disorders/metabolism
14.
Exp Ther Med ; 22(2): 851, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34149897

ABSTRACT

Prostate cancer (PCa) is one of the most common types of cancer and is a serious threat to men's health due to the high rate of incidence and metastasis. However, the exact underlying pathology of this malignant disease has yet to be fully elucidated. The ezrin-radixin-moesin (ERM) family of proteins are associated with the development and metastasis of various types of cancer. Serine threonine kinase 10 (STK10) is an ERM kinase that is involved in the activation of ERM proteins and serves essential roles in the aggregation and adhesion of lymphocytes. To evaluate the functional roles of STK10 in the pathogenesis of PCa, a STK10-knockout (KO) DU145 PCa cell line was generated using the CRISPR-Cas9 gene editing system, and the effects of STK10 deletion on tumor biological behaviors were further analyzed. The present data suggested that STK10 KO promoted PCa cell proliferation by inhibiting p38 MAPK activation and suppressed migration primarily via the inhibition of p38 MAPK signaling and ERM protein activation. To the best of our knowledge, this is the first study to provide evidence that STK10 plays important roles in the proliferation and migration of PCa cells, which will be useful for further investigation into the pathogenesis of this disease.

15.
Cell Death Dis ; 12(4): 362, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33824276

ABSTRACT

Adhesion G protein-coupled receptor A1 (ADGRA1, also known as GPR123) belongs to the G protein-coupled receptors (GPCRs) family and is well conserved in the vertebrate lineage. However, the structure of ADGRA1 is unique and its physiological function remains unknown. Previous studies have shown that Adgra1 is predominantly expressed in the central nervous system (CNS), indicating its important role in the transduction of neural signals. The aim of this study is to investigate the central function of Adgra1 in vivo and clarify its physiological significance by establishing an Adgra1-deficient mouse (Adgra1-/-) model. The results show that Adgra1-/- male mice exhibit decreased body weight with normal food intake and locomotion, shrinkage of body mass, increased lipolysis, and hypermetabolic activity. Meanwhile, mutant male mice present elevated core temperature coupled with resistance to hypothermia upon cold stimulus. Further studies show that tyrosine hydroxylase (TH) and ß3-adrenergic receptor (ß3-AR), indicators of sympathetic nerve excitability, are activated as well as their downstream molecules including uncoupling protein 1 (UCP1), coactivator 1 alpha (PGC1-α) in brown adipose tissue (BAT), and hormone-sensitive lipase (HSL) in white adipose tissue (WAT). In addition, mutant male mice have higher levels of serum T3, T4, accompanied by increased mRNAs of hypothalamus-pituitary-thyroid axis. Finally, Adgra1-/- male mice present abnormal activation of PI3K/AKT/GSK3ß and MEK/ERK pathways in hypothalamus. Overexpression of ADGRA1 in Neuro2A cell line appears to suppress these two signaling pathways. In contrast, Adgra1-/- female mice show comparable body weight along with normal metabolic process to their sex-matched controls. Collectively, ADGRA1 is a negative regulator of sympathetic nervous system (SNS) and hypothalamus-pituitary-thyroid axis by regulating PI3K/AKT/GSK3ß and MEK/ERK pathways in hypothalamus of male mice, suggesting an important role of ADGRA1 in maintaining metabolic homeostasis including energy expenditure and thermogenic balance.


Subject(s)
Adipose Tissue, White/metabolism , Hypothalamus/metabolism , Receptors, G-Protein-Coupled/metabolism , Thermogenesis/physiology , Adipose Tissue, Brown/metabolism , Animals , Energy Metabolism/physiology , Male , Mice , Obesity/metabolism , Signal Transduction/physiology , Sympathetic Nervous System/metabolism , Thyroid Gland/metabolism
16.
Transl Cancer Res ; 9(11): 7079-7090, 2020 Nov.
Article in English | MEDLINE | ID: mdl-35117313

ABSTRACT

BACKGROUND: Serine threonine kinase 10 (STK10) is an ERM kinase involved in the activation of ERM proteins and plays an essential role in the aggregation and adhesion of lymphocytes. STK10 is expressed in about 17 cancer types, including cervical cancer. Cervical cancer is the fourth most common cancer that seriously threatens women's health worldwide. Previous studies have shown that STK10 may affect LFA-1-mediated cell adhesion. Other studies reported a mutation (R634H) of STK10 detected in peripheral T-cell lymphoma. This study aimed to evaluate the functional roles of STK10 in the pathogenesis of cervical cancer. METHODS: We generated STK10 knockout cervical cancer cell lines using the CRISPR-Cas9 gene-editing system, and further analyzed the effects of STK10 deficiency on tumor biological behaviors. The proliferation, apoptosis, migration and invasive activity of these cells were respectively detected by BrdU incorporation, AnnexinV/propidium iodide (PI) staining, wound healing assay and Transwell assays without and with Matrigel. The phosphorylation and expression level of indicated proteins were analyzed by Western blot. The differential expression genes between STK10 knockout and control cells were identified by RNA-seq analysis and further confirmed using qRT-PCR. RESULTS: Our data revealed that target deletion of STK10 does not affect cell proliferation and apoptosis, but promotes the adhesion, migration, and invasion of cervical cancer cells. Most strikingly, the phosphorylation and expression level of ezrin and other ERM proteins in STK10 knockout cells was comparable with that in the control cells. Further, RNA-seq analysis indicated that the knockout of STK10 resulted in a profound alteration of gene expression in cervical cancer cells. CONCLUSIONS: This is the first study to provide evidence that STK10 executes various physiological functions in addition to phosphorylation of ERM proteins, and plays a vital role in the migration and invasion of cervical cancer cells.

17.
Acta Neuropathol Commun ; 8(1): 32, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32169121

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is a group of inherited neurological disorders of the peripheral nervous system. CMT is subdivided into two main types: a demyelinating form, known as CMT1, and an axonal form, known as CMT2. Nearly 30 genes have been identified as a cause of CMT2. One of these is the 'dehydrogenase E1 and transketolase domain containing 1' (DHTKD1) gene. We previously demonstrated that a nonsense mutation [c.1455 T > G (p.Y485*)] in exon 8 of DHTKD1 is one of the disease-causing mutations in CMT2Q (MIM 615025). The aim of the current study was to investigate whether human disease-causing mutations in the Dhtkd1 gene cause CMT2Q phenotypes in a mouse model in order to investigate the physiological function and pathogenic mechanisms associated with mutations in the Dhtkd1 gene in vivo. Therefore, we generated a knock-in mouse model with the Dhtkd1Y486* point mutation. We observed that the Dhtkd1 expression level in sciatic nerve of knock-in mice was significantly lower than in wild-type mice. Moreover, a histopathological phenotype was observed, reminiscent of a peripheral neuropathy, including reduced large axon diameter and abnormal myelination in peripheral nerves. The knock-in mice also displayed clear sensory defects, while no abnormalities in the motor performance were observed. In addition, accumulation of mitochondria and an elevated energy metabolic state was observed in the knock-in mice. Taken together, our study indicates that the Dhtkd1Y486* knock-in mice partially recapitulate the clinical phenotypes of CMT2Q patients and we hypothesize that there might be a compensatory effect from the elevated metabolic state in the knock-in mice that enables them to maintain their normal locomotor function.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Ketoglutarate Dehydrogenase Complex/genetics , Mice , Mitochondria/pathology , Sciatic Nerve/metabolism , Somatosensory Disorders/genetics , Animals , Axons/pathology , Axons/ultrastructure , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/physiopathology , Codon, Nonsense , Energy Metabolism , Gene Knock-In Techniques , Ketoglutarate Dehydrogenase Complex/metabolism , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Neural Conduction , Nonsense Mediated mRNA Decay/genetics , Peripheral Nerves/pathology , Peripheral Nerves/ultrastructure , Phenotype , Point Mutation , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , Somatosensory Disorders/pathology , Somatosensory Disorders/physiopathology
18.
Lab Invest ; 89(9): 1043-52, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564844

ABSTRACT

Liver regeneration is a very complex and well-orchestrated process associated with signaling cascades involving cytokines, growth factors, and metabolic pathways. Adiponectin is an adipocytokine secreted by mature adipocytes, and its receptors are widely distributed in many tissues, including the liver. Adiponectin has direct actions in the liver with prominent roles to improve hepatic insulin sensitivity, increase fatty acid oxidation, and decrease inflammation. To test the hypothesis that adiponectin is required for normal progress of liver regeneration, 2/3 partial hepatectomy (PH) was performed on wild-type and adiponectin-null mice. Compared to wild-type mice, adiponectin-null mice displayed decreased liver mass regrowth, impeded hepatocyte proliferation, and increased hepatic lipid accumulation. Gene expression analysis revealed that adiponectin regulated the gene transcription related to lipid metabolism. Furthermore, the suppressed hepatocyte proliferation was accompanied with reduced signal transducer and activator of transcription protein 3 (STAT3) activity and enhanced suppressor of cytokine signaling 3 (Socs3) transcription. In conclusion, adiponectin-null mice exhibit impaired liver regeneration and increased hepatic steatosis. Increased expression of Socs3 and subsequently reduced activation of STAT3 in adiponectin-null mice may contribute to the alteration of the liver regeneration capability and hepatic lipid metabolism after PH.


Subject(s)
Adiponectin/deficiency , Liver Regeneration/physiology , STAT3 Transcription Factor/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Hepatectomy , Hepatocytes/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Liver/surgery , Male , Mice , Mice, Knockout , Phosphorylation , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription, Genetic , Transcriptional Activation
19.
J Endocrinol ; 243(2): 111-123, 2019 11.
Article in English | MEDLINE | ID: mdl-31454789

ABSTRACT

Obesity and type 2 diabetes (T2D) are both complicated endocrine disorders resulting from an interaction between multiple predisposing genes and environmental triggers, while diet and exercise have key influence on metabolic disorders. Previous reports demonstrated that 2-aminoadipic acid (2-AAA), an intermediate metabolite of lysine metabolism, could modulate insulin secretion and predict T2D, suggesting the role of 2-AAA in glycolipid metabolism. Here, we showed that treatment of diet-induced obesity (DIO) mice with 2-AAA significantly reduced body weight, decreased fat accumulation and lowered fasting glucose. Furthermore, Dhtkd1-/- mice, in which the substrate of DHTKD1 2-AAA increased to a significant high level, were resistant to DIO and obesity-related insulin resistance. Further study showed that 2-AAA induced higher energy expenditure due to increased adipocyte thermogenesis via upregulating PGC1α and UCP1 mediated by ß3AR activation, and stimulated lipolysis depending on enhanced expression of hormone-sensitive lipase (HSL) through activating ß3AR signaling. Moreover, 2-AAA could alleviate the diabetic symptoms of db/db mice. Our data showed that 2-AAA played an important role in regulating glycolipid metabolism independent of diet and exercise, implying that improving the level of 2-AAA in vivo could be developed as a strategy in the treatment of obesity or diabetes.


Subject(s)
2-Aminoadipic Acid/pharmacology , Body Weight/drug effects , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , 2-Aminoadipic Acid/metabolism , 3T3-L1 Cells , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat/adverse effects , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/physiopathology , Protective Agents/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction/drug effects , Thermogenesis/drug effects
20.
J Neurosci Res ; 86(2): 465-70, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17918738

ABSTRACT

gamma-Aminobutyric acid (GABA) transporters play a key role in the regulation of GABA neurotransmission. We reported previously that overexpression of the GABA transporter subtype 1 (GAT1), the major form of the GABA transporter in the CNS, led to hyperalgesia in mice. In the present study, nociceptive responses of GAT1-knockout mice (GAT1(-/-)) were compared with those of heterozygous (GAT(+/-)) and wild-type (GAT(+/+)) mice by four conventional pain models (tail-immersion test, hot-plate test, acetic acid-induced abdominal constriction test, and formalin test). In addition, the analgesic effects of two GAT1-selective inhibitors, NO-711 and tiagabine, were examined in all three genotypes using the same four models. Our data demonstrated that GAT1 deficiency because of genetic knockout or acute blockade by selective inhibitors leads to hypoalgesia in mice. These results confirmed the crucial role of GAT1 in the regulation of nociceptive threshold and suggested that GAT1 inhibitors have the potential for clinical use in pain therapy.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Pain Threshold/physiology , Pain/physiopathology , Animals , GABA Antagonists/pharmacology , GABA Plasma Membrane Transport Proteins/drug effects , GABA Plasma Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Nipecotic Acids/pharmacology , Oximes/pharmacology , Pain Threshold/drug effects , Tiagabine
SELECTION OF CITATIONS
SEARCH DETAIL