Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 623
Filter
1.
Pharmacology ; 109(2): 98-109, 2024.
Article in English | MEDLINE | ID: mdl-38325349

ABSTRACT

INTRODUCTION: Membrane-associated guanylate kinase with an inverted domain structure-1 (MAGI1) is dysregulated in diabetes; however, its role in diabetic nephropathy (DN) remains unclear. In this study, we determined the function and associated mechanisms of MAGI1 in DN. METHODS: Serum samples from 28 patients with DN and 28 normal volunteers were collected. High-glucose (HG)-treated human renal mesangial cells (HRMCs) and streptozotocin-treated rats were used as cell and animal models of DN, respectively. MAGI1 mRNA expression was measured by quantitative reverse transcription polymerase chain reaction. An 5-Ethynyl-2'-deoxyuridine assay was used to assess cell proliferation, whereas Western blot analysis was performed to quantitate the levels of markers associated with proliferation, the extracellular matrix (ECM), and inflammation. These included collagens I, collagen IV, cyclin D1, AKT, phosphorylated-AKT (p-AKT), PI3K, and phosphorylated-PI3K (p-PI3K). The predicted binding of miR-205-5p with the MAGI1 3'UTR was verified using a luciferase assay. RESULTS: MAGI1 expression was increased in serum samples from DN patients and in HRMCs treated with HG. MAGI1 knockdown attenuated excessive proliferation, ECM accumulation, and inflammation in HG-induced HRMCs as well as injury to DN rats. MiR-205-5p potentially interacted with the 3'UTR of MAGI1 and binding was verified using a dual-luciferase reporter assay. Moreover, miR-205-5p repression offset the inhibitory influence of MAGI1 knockdown on proliferation, collagen deposition, and inflammation in HG-treated HRMCs. CONCLUSION: MAGI1 contributes to injury caused by DN. Furthermore, miR-205-5p binds to MAGI1 and suppresses MAGI1 function. These findings suggest that miR-205-5p-mediates MAGI1 inhibition, which represents a potential treatment for DN.


Subject(s)
Diabetic Nephropathies , MicroRNAs , Animals , Humans , Rats , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Collagen/metabolism , Diabetes Mellitus , Diabetic Nephropathies/genetics , Glucose/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Inflammation/genetics , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
2.
Glia ; 72(5): 857-871, 2024 May.
Article in English | MEDLINE | ID: mdl-38234042

ABSTRACT

Tumor-associated astrocytes (TAAs) in the glioblastoma microenvironment play an important role in tumor development and malignant progression initiated by glioma stem cells (GSCs). In the current study, normal human astrocytes (NHAs) were cultured and continuously treated with GSC-derived exosomes (GSC-EXOs) induction to explore the mechanism by which GSCs affect astrocyte remodeling. This study revealed that GSC-EXOs can induce the transformation of NHAs into TAAs, with relatively swollen cell bodies and multiple extended processes. In addition, high proliferation, elevated resistance to temozolomide (TMZ), and increased expression of TAA-related markers (TGF-ß, CD44, and tenascin-C) were observed in the TAAs. Furthermore, GSC-derived exosomal miR-3065-5p could be delivered to NHAs, and miR-3065-5p levels increased significantly in TAAs, as verified by miRNA expression profile sequencing and Reverse transcription polymerase chain reaction. Overexpression of miR-3065-5p also enhanced NHA proliferation, elevated resistance to TMZ, and increased the expression levels of TAA-related markers. In addition, both GSC-EXO-induced and miR-3065-5p-overexpressing NHAs promoted tumorigenesis of GSCs in vivo. Discs Large Homolog 2 (DLG2, downregulated in glioblastoma) is a direct downstream target of miR-3065-5p in TAAs, and DLG2 overexpression could partially reverse the transformation of NHAs into TAAs. Collectively, these data demonstrate that GSC-EXOs induce the transformation of NHAs into TAAs via the miR-3065-5p/DLG2 signaling axis and that TAAs can further promote the tumorigenesis of GSCs. Thus, precisely blocking the interactions between astrocytes and GSCs via exosomes may be a novel strategy to inhibit glioblastoma development, but more in-depth mechanistic studies are still needed.


Subject(s)
Exosomes , Glioblastoma , Glioma , MicroRNAs , Humans , Glioblastoma/pathology , Astrocytes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Exosomes/metabolism , Glioma/pathology , Temozolomide/pharmacology , Temozolomide/metabolism , Neoplastic Stem Cells/metabolism , Carcinogenesis/genetics , Cell Proliferation , Tumor Microenvironment , Tumor Suppressor Proteins/metabolism , Guanylate Kinases/metabolism
3.
Cell Signal ; 113: 110954, 2024 01.
Article in English | MEDLINE | ID: mdl-38084836

ABSTRACT

Revealing the role of non-coding RNAs (ncRNAs) in inducing dysregulated pathological responses to external signals may identify therapeutic targets for inhibiting the progression of clear cell renal cell carcinoma (ccRCC). Non-coding RNAs belong to a class of RNA molecules that do not encode proteins but possess diverse biological functions, playing essential roles in the occurrence and development of metastatic and proliferative tumors. To investigate the impact of the upstream interaction between miR-142-3p and lncRNA MAGI2-AS3 on the tumor-suppressive activity of the STAM gene, we firstly conducted bioinformatics analysis to predict the upstream miRNAs of STAM and the upstream lncRNAs of the miRNAs through online databases (miRanda, miRDB, TargetScan, LncBase v2), which were further validated by the starBasev2.0 database. Subsequently, multiple experimental techniques were employed to validate these findings, including RT-qPCR, Western blotting, measurement of cellular functional activity, and luciferase reporter assays. Through these experimental methods, we provided compelling evidence regarding the role of miR-142-3p and MAGI2-AS3 in regulating STAM gene expression and functionality, revealing their potential significance in tumor suppression. Our research demonstrates the importance of the MAGI2-AS3/miR-142-3p/STAM signaling pathway axis in ccRCC. MAGI2-AS3 competes for binding with miR-142-3p, resulting in upregulated STAM gene expression. This upregulation inhibits tumor proliferation and metastasis in ccRCC cells. Conversely, overexpression of miR-142-3p or silencing of MAGI2-AS3 promotes tumor behavior, while downregulation of miR-142-3p inhibits the development of ccRCC. Targeting the MAGI2-AS3/miR-142-3p/STAM axis holds promise as a therapeutic strategy for ccRCC treatment.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Kidney Neoplasms/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism
4.
J Biol Chem ; 300(1): 105575, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38110034

ABSTRACT

The carboxy-terminal tail of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) envelope protein (E) contains a PDZ-binding motif (PBM) which is crucial for coronavirus pathogenicity. During SARS-CoV-2 infection, the viral E protein is expressed within the Golgi apparatus membrane of host cells with its PBM facing the cytoplasm. In this work, we study the molecular mechanisms controlling the presentation of the PBM to host PDZ (PSD-95/Dlg/ZO-1) domain-containing proteins. We show that at the level of the Golgi apparatus, the PDZ-binding motif of the E protein is not detected by E C-terminal specific antibodies nor by the PDZ domain-containing protein-binding partner. Four alanine substitutions upstream of the PBM in the central region of the E protein tail is sufficient to generate immunodetection by anti-E antibodies and trigger robust recruitment of the PDZ domain-containing protein into the Golgi organelle. Overall, this work suggests that the presentation of the PBM to the cytoplasm is under conformational regulation mediated by the central region of the E protein tail and that PBM presentation probably does not occur at the surface of Golgi cisternae but likely at post-Golgi stages of the viral cycle.


Subject(s)
Coronavirus Envelope Proteins , Cytoplasm , SARS-CoV-2 , Humans , Amino Acid Motifs , Coronavirus Envelope Proteins/chemistry , Coronavirus Envelope Proteins/metabolism , COVID-19/pathology , COVID-19/virology , Cytoplasm/metabolism , Cytoplasm/virology , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Guanylate Kinases/metabolism , PDZ Domains , Protein Binding , Protein Conformation , Protein Transport , SARS-CoV-2/chemistry , SARS-CoV-2/metabolism
5.
Sci Rep ; 13(1): 22027, 2023 12 12.
Article in English | MEDLINE | ID: mdl-38086879

ABSTRACT

Brain-enriched guanylate kinase-associated protein (BEGAIN) is highly enriched in the post-synaptic density (PSD) fraction and was identified in our previous study as a protein associated with neuropathic pain in the spinal dorsal horn. PSD protein complexes containing N-methyl-D-aspartate receptors are known to be involved in neuropathic pain. Since these PSD proteins also participate in learning and memory, BEGAIN is also expected to play a crucial role in this behavior. To verify this, we first examined the distribution of BEGAIN in the brain. We found that BEGAIN was widely distributed in the brain and highly expressed in the dendritic regions of the hippocampus. Moreover, we found that BEGAIN was concentrated in the PSD fraction of the hippocampus. Furthermore, immunoelectron microscopy confirmed that BEGAIN was localized at the asymmetric synapses. Behavioral tests were performed using BEGAIN-knockout (KO) mice to determine the contribution of BEGAIN toward learning and memory. Spatial reference memory and reversal learning in the Barns circular maze test along with contextual fear and cued fear memory in the contextual and cued fear conditioning test were significantly impaired in BEGAIN-KO mice compared to with those in wild-type mice. Thus, this study reveals that BEGAIN is a component of the post-synaptic compartment of excitatory synapses involved in learning and memory.


Subject(s)
Neuralgia , Post-Synaptic Density , Mice , Animals , Post-Synaptic Density/metabolism , Brain/metabolism , Hippocampus/metabolism , Mice, Knockout , Maze Learning , Guanylate Kinases/metabolism , Neuralgia/metabolism
6.
Drug Dev Res ; 84(7): 1482-1495, 2023 11.
Article in English | MEDLINE | ID: mdl-37551766

ABSTRACT

Drug resistance is a major impediment to the successful treatment of glioma. This study aimed to elucidate the effects and mechanisms of the long noncoding RNA membrane-associated guanylate kinase inverted-2 antisense RNA 3 (MAGI2-AS3) on temozolomide (TMZ) resistance in glioma cells. MAGI2-AS3 expression in TMZ-resistant glioblastoma (GBM) cells was analyzed using the Gene Expression Omnibus data set GSE113510 and quantitative real-time PCR (qRT-PCR). Cell viability and TMZ half-maximal inhibitory concentration values were determined using the MTT assay. Apoptosis and cell cycle distribution were evaluated using flow cytometry. The expression of multidrug resistance 1 (MDR1), ATP-binding cassette superfamily G member 2 (ABCG2), protein kinase B (Akt), and phosphorylated Akt was detected using qRT-PCR and/or western blot analysis. MAGI2-AS3 was expressed at low levels in TMZ-resistant GBM cells relative to that in their parental cells. MAGI2-AS3 re-expression alleviated TMZ resistance in TMZ-resistant GBM cells. MAGI2-AS3 overexpression also accelerated TMZ-induced apoptosis and G2/M phase arrest. Mechanistically, MAGI2-AS3 overexpression reduced MDR1 and ABCG2 expression and inhibited the Akt pathway, whereas Akt overexpression abrogated the reduction in MDR1 and ABCG2 expression induced by MAGI2-AS3. Moreover, activation of the Akt pathway inhibited the effects of MAGI2-AS3 on TMZ resistance. MAGI2-AS3 inhibited tumor growth and enhanced the suppressive effect of TMZ on glioma tumorigenesis in vivo. In conclusion, MAGI2-AS3 reverses TMZ resistance in glioma cells by inactivating the Akt pathway.


Subject(s)
Glioblastoma , Glioma , MicroRNAs , RNA, Long Noncoding , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , Proto-Oncogene Proteins c-akt/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , RNA, Antisense/pharmacology , RNA, Antisense/therapeutic use , Cell Line, Tumor , Cell Proliferation , MicroRNAs/genetics , Drug Resistance, Neoplasm/genetics , RNA, Long Noncoding/genetics , Gene Expression Regulation, Neoplastic , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Guanylate Kinases/pharmacology
7.
Int J Biol Sci ; 19(10): 3099-3114, 2023.
Article in English | MEDLINE | ID: mdl-37416779

ABSTRACT

Background: Chemotherapy resistance is a significant cause for poor prognosis of epithelial ovarian cancer (EOC). However, the molecular mechanism of chemo-resistance remains unclear, and developing available therapies and effective biomarkers for resistant EOC is in urgent demand. Stemness of cancer cells directly results in chemo-resistance. Exosomal miRNAs rebuild tumor microenvironment (TME) and act as widely used clinical liquid biopsy markers. Methods: In our study, high throughput screenings and comprehensive analysis were performed to screen for miRNAs, which were both up-regulated in resistant EOC tissues and related to stemness, and miR-6836 was identified accordingly. Results: Clinically, high miR-6836 expression was closely correlated with poor chemotherapy response and survival for EOC patients. Functionally, miR-6836 promoted EOC cell cisplatin resistance by increasing stemness and suppressing apoptosis. Mechanistically, miR-6836 directly targeted DLG2 to enhance Yap1 nuclear translocation, and was regulated by TEAD1 forming the positive feedback loop: miR-6836-DLG2-Yap1-TEAD1. Furthermore, miR-6836 could be packaged into secreted exosomes in cisplatin-resistant EOC cells and exosomal miR-6836 was able to be delivered into cisplatin-sensitive EOC cells and reverse their cisplatin response. Conclusion: Our study revealed the molecular mechanisms of chemotherapy resistance, and identified miR-6836 as the possible therapeutic target and effective biopsy marker for resistant EOC.


Subject(s)
Exosomes , MicroRNAs , Ovarian Neoplasms , Humans , Female , Cisplatin/pharmacology , Cisplatin/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , MicroRNAs/metabolism , Signal Transduction/genetics , Exosomes/genetics , Exosomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Tumor Microenvironment , Tumor Suppressor Proteins/metabolism , Guanylate Kinases/metabolism
8.
J Cell Sci ; 136(11)2023 06 01.
Article in English | MEDLINE | ID: mdl-37288673

ABSTRACT

Gap junction channels, composed of connexins, allow direct cell-to-cell communication. Connexin 43 (Cx43; also known as GJA1) is widely expressed in tissues, including the epidermis. In a previous study of human papillomavirus-positive cervical epithelial tumour cells, we identified Cx43 as a binding partner of the human homologue of Drosophila Discs large (Dlg1; also known as SAP97). Dlg1 is a member of the membrane associated-guanylate kinase (MAGUK) scaffolding protein family, which is known to control cell shape and polarity. Here, we show that Cx43 also interacts with Dlg1 in uninfected keratinocytes in vitro and in keratinocytes, dermal cells and adipocytes in normal human epidermis in vivo. Depletion of Dlg1 in keratinocytes did not alter Cx43 transcription but was associated with a reduction in Cx43 protein levels. Reduced Dlg1 levels in keratinocytes resulted in a reduction in Cx43 at the plasma membrane with a concomitant reduction in gap junctional intercellular communication and relocation of Cx43 to the Golgi compartment. Our data suggest a key role for Dlg1 in maintaining Cx43 at the plasma membrane in keratinocytes.


Subject(s)
Connexin 43 , Discs Large Homolog 1 Protein , Keratinocytes , Humans , Cell Communication , Cell Membrane/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Gap Junctions/metabolism , Guanylate Kinases/metabolism , Keratinocytes/metabolism , Discs Large Homolog 1 Protein/genetics , Discs Large Homolog 1 Protein/metabolism
9.
Sci Adv ; 9(19): eadf3295, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37163606

ABSTRACT

Dynamic signal transduction requires the rapid assembly and disassembly of signaling complexes, often mediated by phosphoprotein binding modules. The guanylate kinase-like (GK) domain of the membrane-associated guanylate kinases (MAGUKs) is such a module orchestrating signaling at cellular junctions. The MAGI subfamily of MAGUKs contains a truncated GK domain with unknown structure and function, although they participate in diverse physiological and pathological processes. Here, we demonstrate that the truncated GK domain of MAGI2 interacts with its adjacent PDZ0 domain to form a structural supramodule capable of recognizing phosphoproteins. A conserved phosphorylation-dependent binding motif for PDZ0-GK is delineated, which leads to identification of a set of previously unknown binding partners. We explore the structure and function of the MAGI2-target complex with an inhibitory peptide derived from the consensus motif. Our work reveals an action mechanism of the cryptic MAGI GKs and broadens our understanding of the target recognition rules of phosphoprotein binding modules.


Subject(s)
Phosphoproteins , Guanylate Kinases/genetics , Guanylate Kinases/chemistry , Guanylate Kinases/metabolism , Phosphorylation , Amino Acid Sequence , Protein Binding , Phosphoproteins/metabolism
10.
Cells ; 12(8)2023 04 18.
Article in English | MEDLINE | ID: mdl-37190086

ABSTRACT

Microcephaly with pontine and cerebellar hypoplasia (MICPCH) syndrome is a neurodevelopmental disorder caused by the deficiency of the X-chromosomal gene CASK. However, the molecular mechanisms by which CASK deficiency causes cerebellar hypoplasia in this syndrome remain elusive. In this study, we used CASK knockout (KO) mice as models for MICPCH syndrome and investigated the effect of CASK mutants. Female CASK heterozygote KO mice replicate the progressive cerebellar hypoplasia observed in MICPCH syndrome. CASK KO cultured cerebellar granule (CG) cells show progressive cell death that can be rescued by co-infection with lentivirus expressing wild-type CASK. Rescue experiments with CASK deletion mutants identify that the CaMK, PDZ, and SH3, but not L27 and guanylate kinase domains of CASK are required for the survival of CG cells. We identify missense mutations in the CaMK domain of CASK derived from human patients that fail to rescue the cell death of cultured CASK KO CG cells. Machine learning-based structural analysis using AlphaFold 2.2 predicts that these mutations disrupt the structure of the binding interface with Liprin-α2. These results suggest that the interaction with Liprin-α2 via the CaMK domain of CASK may be involved in the pathophysiology of cerebellar hypoplasia in MICPCH syndrome.


Subject(s)
Adaptor Proteins, Signal Transducing , Cerebellum , Guanylate Kinases , Membrane Proteins , Mental Retardation, X-Linked , Microcephaly , Cerebellum/metabolism , Cerebellum/pathology , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Guanylate Kinases/chemistry , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Humans , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Mice, Knockout , Animals , Mice , Female , Cells, Cultured , Mutation , Protein Domains , Machine Learning , Software , Apoptosis
11.
Pathol Res Pract ; 246: 154530, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37196468

ABSTRACT

MAGI2 Antisense RNA 3 (MAGI2-AS3) is a long non-coding RNA (lncRNA) transcribed from a locus on 7q21.11. This lncRNA has been described to be abnormally expressed in a variety of malignancies in correlation with many clinical characteristics. Moreover, it might participate in the pathogenesis of congenital diaphragmatic hernia, Alzheimer's disease and intervertebral disc degeneration. Mechanistically, MAGI2-AS3 can serve as a molecular sponge for miR-142-3p, miR-424-5p, miR-15b, miR-233, miR-452-5p, miR-629-5p, miR-25, miR-155, miR-23a-3p, miR-519c-3p, miR-374b-5p, miR-374a, miR-31-5p, miR-3163, miR-525-5p, miR-15-5p, miR-374a-5p, miR-374b-5p, miR-218-5p, miR-141-3p and miR-200a-3p to regulate expression of their mRNA targets. The current review summarizes the role of MAGI2-AS3 in different disorders to highlight its importance in their pathophysiology.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Humans , Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation/genetics , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Antisense
12.
Mol Cell Neurosci ; 125: 103856, 2023 06.
Article in English | MEDLINE | ID: mdl-37105372

ABSTRACT

In the central nervous system, glutamatergic synapses play a central role in the regulation of excitatory neuronal transmission. With the membrane-associated guanylate kinase (MAGUK) family of proteins as their structuring scaffold, glutamatergic receptors serve as the powerhouse of glutamatergic synapses. Glutamatergic receptors can be categorized as metabotropic and ionotropic receptors. The latter are then categorized into N-methyl-d-aspartate, kainate receptors, and α-amino-3-hydroxy-5-methyl-isoxazole-propionic acid receptors (AMPARs). Over the past two decades, genetic tagging technology and super-resolution microscopy have been of the utmost importance to unravel how the different receptors are organized at glutamatergic synapses. At the plasma membrane, receptors are highly mobile but show reduced mobility when at synaptic sites. This partial immobilization of receptors at synaptic sites is attributed to the stabilization/anchoring of receptors with the postsynaptic MAGUK proteins and auxiliary proteins, and presynaptic proteins. These partial immobilizations and localization of glutamatergic receptors within the synaptic sites are fundamental for proper basal transmission and synaptic plasticity. Perturbations of the stabilization of glutamatergic receptors are often associated with cognitive deficits. In this review, we describe the proposed mechanisms for synaptic localization and stabilization of AMPARs, the major players of fast excitatory transmission in the central nervous system.


Subject(s)
Receptors, AMPA , Synapses , Receptors, AMPA/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Neuronal Plasticity/physiology , Guanylate Kinases/metabolism
13.
Immun Inflamm Dis ; 11(4): e772, 2023 04.
Article in English | MEDLINE | ID: mdl-37102649

ABSTRACT

BACKGROUND: Intervertebral disc degeneration (IDD) is a pathological process that occurs during the natural aging of intervertebral discs. Accumulating evidence suggests that noncoding RNAs (ncRNAs), including microRNAs and long ncRNAs (lncRNAs), participate in the pathogenesis and development of IDD. Herein, we examined the role of lncRNA MAGI2-AS3 in the pathogenic mechanism of IDD. MATERIAL AND METHODS: To develop an IDD in vitro model, we treated human nucleus pulposus (NP) cells with lipopolysaccharide (LPS). Aberrant levels of lncRNA MAGI2-AS3, miR-374b-5p, interleukin (IL)-10 and extracellular matrix (ECM)-related proteins in NP cells were examined using reverse transcription-quantitative PCR and western blot analysis. LPS-induced NP cell injury and inflammatory response were confirmed using the MTT assay, flow cytometry, Caspase3 activity, and enzyme-linked immunosorbent assay. Dual-luciferase reporter assay and rescue experiments were performed to confirm targets between lncRNA MAGI2-AS3 and miR-374b-5p or miR-374b-5p and IL-10. RESULTS: LPS-induced NP cells exhibited low levels of lncRNA MAGI2-AS3 and IL-10 expression, along with high miR-374b-5p expression. miR-374b-5p was a target of lncRNA MAGI2-AS3 and IL-10. LncRNA MAGI2-AS3 ameliorated injury, inflammatory response, and ECM degradation in LPS-treated NP cells by downregulating miR-374b-5p to upregulate IL-10 expression. CONCLUSIONS: LncRNA MAGI2-AS3 increased IL-10 expression levels by sponging miR-374b-5p, which, in turn, alleviated LPS-triggered decreased NP cell proliferation and increased apoptosis, inflammatory response, and ECM degradation. Therefore, lncRNA MAGI2-AS3 may be a potential therapeutic target for IDD.


Subject(s)
Intervertebral Disc Degeneration , MicroRNAs , Nucleus Pulposus , RNA, Long Noncoding , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins , Guanylate Kinases/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukins , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Lipopolysaccharides/toxicity , MicroRNAs/genetics , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
14.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37115584

ABSTRACT

Hematopoietic stem cells (HSC) and downstream lineage-biased multipotent progenitors (MPP) tailor blood production and control myelopoiesis on demand. Recent lineage tracing analyses revealed MPPs to be major functional contributors to steady-state hematopoiesis. However, we still lack a precise resolution of myeloid differentiation trajectories and cellular heterogeneity in the MPP compartment. Here, we found that myeloid-biased MPP3 are functionally and molecularly heterogeneous, with a distinct subset of myeloid-primed secretory cells with high endoplasmic reticulum (ER) volume and FcγR expression. We show that FcγR+/ERhigh MPP3 are a transitional population serving as a reservoir for rapid production of granulocyte/macrophage progenitors (GMP), which directly amplify myelopoiesis through inflammation-triggered secretion of cytokines in the local bone marrow (BM) microenvironment. Our results identify a novel regulatory function for a secretory MPP3 subset that controls myeloid differentiation through lineage-priming and cytokine production and acts as a self-reinforcing amplification compartment in inflammatory stress and disease conditions.


Subject(s)
Hematopoiesis , Receptors, IgG , Cell Differentiation , Cell Lineage , Myeloid Cells , Guanylate Kinases/metabolism , Membrane Proteins/metabolism
15.
ACS Chem Neurosci ; 14(6): 1107-1118, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36878000

ABSTRACT

Multiple sclerosis (MS) is a chronic disease and one of the leading causes of disability in young adults. The current study aims to investigate the pathogenesis of MS via studying the regulatory role of novel lncRNA MAGI2-AS3 in miR-374b-5p and their downstream targets PTEN/AKT/IRF-3/IFN-ß and the relationship of this pathway with disease severity. Moreover, it aims to assess the role of MAGI2-AS3/miR-374b-5p as diagnostic and/or prognostic biomarkers for MS. Overall, 150 contributors were recruited: 100 patients with MS and 50 healthy volunteers. Gene expression of MAGI2-AS3, miR-374b-5p, PTEN, AKT, and IRF-3 were assessed using RT-qPCR, and IFN-ß was measured by ELISA. Compared with the healthy control group, serum MAGI2-AS3 and PTEN were downregulated in MS patients, whereas miR-374b-5p, PI3K, AKT, IRF-3, and IFN-ß were upregulated in MS patients. Furthermore, MAGI2-AS3 was downregulated, while miR-374b-5p was upregulated in MS patients with an expanded disability status scale (EDSS) ≥3.5, compared to patients with an EDSS <3.5. Receiver-operating-characteristic curve analysis revealed that MAGI2-AS3 and miR-374b-5p can be used in the diagnosis of MS. Remarkably, multivariate logistic analysis revealed that MAGI2-AS3, miR-374b-5p, PTEN, and AKT act as independent variables in MS. Moreover, MAGI2-AS3 was directly correlated with PTEN and inversely correlated with miR-374b-5p, AKT, and EDSS. Regarding miR-374b-5p, it was positively correlated with AKT and EDSS. In conclusion, the study showed for the first time that the crosstalk between MAGI2-AS3 and miR-374b-5p could affect the AKT/IRF3/IFN-ß axis in MS. Interestingly, MAGI2-AS3 and miR-374b-5p could be genetic noninvasive biomarkers for MS.


Subject(s)
MicroRNAs , Multiple Sclerosis , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Multiple Sclerosis/genetics , Biomarkers , Cell Proliferation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism
16.
Genomics ; 115(2): 110599, 2023 03.
Article in English | MEDLINE | ID: mdl-36889366

ABSTRACT

Prostate cancer (PCa) is a common malignant cancer in elderly males in Western countries. Whole-genome sequencing confirmed that long non-coding RNAs (lncRNAs) are frequently altered in castration-resistant prostate cancer (CRPC) and promote drug resistance to cancer therapy. Therefore, elucidating the prospective role of lncRNAs in PCa oncogenesis and progression is of remarkable clinical significance. In this study, gene expression in prostate tissues was determined using RNA-sequencing datasets, and the gene diagnostic and prognostic values of CRPC were analyzed using bioinformatics. Further, the expression levels and clinical significance of MAGI2 Antisense RNA 3 (MAGI2-AS3) in PCa clinical specimens were evaluated. The tumor-suppressive activity of MAGI2-AS3 was functionally explored in PCa cell lines and animal xenograft models. MAGI2-AS3 was found to be aberrantly decreased in CRPC and was negatively correlated with Gleason score and lymph node status. Notably, low MAGI2-AS3 expression positively correlated with poorer survival in patients with PCa. The overexpression of MAGI2-AS3 significantly inhibited the proliferation and migration of PCa in vitro and in vivo. Mechanistically, MAGI2-AS3 could play a tumor suppressor function in CRPC through a novel miR-106a-5p/RAB31 regulatory network and could be a target for future cancer therapy.


Subject(s)
MicroRNAs , Prostatic Neoplasms, Castration-Resistant , RNA, Long Noncoding , Male , Animals , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Prognosis , Cell Proliferation/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism
17.
Life Sci Alliance ; 5(10)2022 10.
Article in English | MEDLINE | ID: mdl-36137748

ABSTRACT

CASK is a unique membrane-associated guanylate kinase (MAGUK) because of its Ca2+/calmodulin-dependent kinase (CaMK) domain. We describe four male patients with a severe neurodevelopmental disorder with microcephaly carrying missense variants affecting the CaMK domain. One boy who carried the p.E115K variant and died at an early age showed pontocerebellar hypoplasia (PCH) in addition to microcephaly, thus exhibiting the classical MICPCH phenotype observed in individuals with CASK loss-of-function variants. All four variants selectively weaken the interaction of CASK with Liprin-α2, a component of the presynaptic active zone. Liprin-α proteins form spherical phase-separated condensates, which we observe here in Liprin-α2 overexpressing HEK293T cells. Large Liprin-α2 clusters were also observed in transfected primary-cultured neurons. Cluster formation of Liprin-α2 is reversed in the presence of CASK; this is associated with altered phosphorylation of Liprin-α2. The p.E115K variant fails to interfere with condensate formation. As the individual carrying this variant had the severe MICPCH disorder, we suggest that regulation of Liprin-α2-mediated phase condensate formation is a new functional feature of CASK which must be maintained to prevent PCH.


Subject(s)
Microcephaly , Calmodulin/genetics , Calmodulin/metabolism , Cerebellar Diseases , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , HEK293 Cells , Humans , Male , Mental Retardation, X-Linked , Microcephaly/genetics , Mutation
18.
J Biol Chem ; 298(11): 102476, 2022 11.
Article in English | MEDLINE | ID: mdl-36096201

ABSTRACT

The accumulation of misfolded proteins is a hallmark of aging and many neurodegenerative diseases, making it important to understand how the cellular machinery recognizes and processes such proteins. A key question in this respect is whether misfolded proteins are handled in a similar way regardless of their genetic origin. To approach this question, we compared how three different misfolded proteins, guk1-7, gus1-3, and pro3-1, are handled by the cell. We show that all three are nontoxic, even though highly overexpressed, highlighting their usefulness in analyzing the cellular response to misfolding in the absence of severe stress. We found significant differences between the aggregation and disaggregation behavior of the misfolded proteins. Specifically, gus1-3 formed some aggregates that did not efficiently recruit the protein disaggregase Hsp104 and did not colocalize with the other misfolded reporter proteins. Strikingly, while all three misfolded proteins generally coaggregated and colocalized to specific sites in the cell, disaggregation was notably different; the rate of aggregate clearance of pro3-1 was faster than that of the other misfolded proteins, and its clearance rate was not hindered when pro3-1 colocalized with a slowly resolved misfolded protein. Finally, we observed using super-resolution light microscopy as well as immunogold labeling EM in which both showed an even distribution of the different misfolded proteins within an inclusion, suggesting that misfolding characteristics and remodeling, rather than spatial compartmentalization, allows for differential clearance of these misfolding reporters residing in the same inclusion. Taken together, our results highlight how properties of misfolded proteins can significantly affect processing.


Subject(s)
Neurodegenerative Diseases , Saccharomyces cerevisiae Proteins , Humans , Protein Aggregates , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Neurodegenerative Diseases/metabolism , Protein Folding , Heat-Shock Proteins/metabolism , Guanylate Kinases/metabolism
19.
Horm Metab Res ; 54(11): 754-759, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35944561

ABSTRACT

Prostate cancer is a common male cancer with high morbidity and mortality worldwide. According to current research, the integration of long non-coding RNA (lncRNAs) and microRNA(miRNAs) can be expressed in a variety of cancers and play an important role in diagnosis. Based on this, this study explored the clinical role of lncRNA MAGI2-AS3 (MAGI2-AS3) in prostate cancer. By detecting the expression levels of MAGI2-AS3 and miR-142-3p, the correlation between the MAGI2-AS3 expression and the characteristics of clinical data was analyzed. ROC curve analysis was performed and the area under the ROC curve (AUC) was used to evaluate the diagnostic value of MAGI2-AS3 in distinguishing prostate cancer patients from healthy controls. The function of MAGI2-AS3 in prostate cancer cells was explored through CCK-8 and Transwell assays, and the relationship between MAGI2-AS3 and miR-142-3p was investigated by luciferase activity assay. MAGI2-AS3 has descended expression while miR-142-3p has an ascendant one in prostate cancer serum samples and cells. ROC curve analysis revealed that the AUC was 0.953 for MAGI2-AS3, with a sensitivity of 91.5% and specificity of 84.7%. Overexpression of MAGI2-AS3 in LNCaP and PC3 cells suppressed the biological function of the cell including proliferation capacity, migration level, and invasion. MAGI2-AS3 was considered a diagnostic biomarker for prostate cancer patients and inhibited prostate cancer progression by targeting miR-142-3p.


Subject(s)
MicroRNAs , Prostatic Neoplasms , RNA, Long Noncoding , Humans , Male , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Adaptor Proteins, Signal Transducing/genetics , Guanylate Kinases/genetics , Guanylate Kinases/metabolism
20.
Blood ; 140(13): 1549-1564, 2022 09 29.
Article in English | MEDLINE | ID: mdl-35895897

ABSTRACT

Deep vein thrombosis (DVT) is the third most common cause of cardiovascular mortality. Several studies suggest that DVT occurs at the intersection of dysregulated inflammation and coagulation upon activation of inflammasome and secretion of interleukin 1ß (IL-1ß) in restricted venous flow conditions. Our recent studies showed a signaling adapter protein, Gab2 (Grb2-associated binder 2), plays a crucial role in propagating inflammatory signaling triggered by IL-1ß and other inflammatory mediators in endothelial cells. The present study shows that Gab2 facilitates the assembly of the CBM (CARMA3 [CARD recruited membrane-associated guanylate kinase protein 3]-BCL-10 [B-cell lymphoma 10]-MALT1 [mucosa-associated lymphoid tissue lymphoma translocation protein 1]) signalosome, which mediates the activation of Rho and NF-κB in endothelial cells. Gene silencing of Gab2 or MALT1, the effector signaling molecule in the CBM signalosome, or pharmacological inhibition of MALT1 with a specific inhibitor, mepazine, significantly reduced IL-1ß-induced Rho-dependent exocytosis of P-selectin and von Willebrand factor (VWF) and the subsequent adhesion of neutrophils to endothelial cells. MALT1 inhibition also reduced IL-1ß-induced NF-κB-dependent expression of tissue factor and vascular cell adhesion molecule 1. Consistent with the in vitro data, Gab2 deficiency or pharmacological inhibition of MALT1 suppressed the accumulation of monocytes and neutrophils at the injury site and attenuated venous thrombosis induced by the inferior vena cava ligation-induced stenosis or stasis in mice. Overall, our data reveal a previously unrecognized role of the Gab2-MALT1 axis in thromboinflammation. Targeting the Gab2-MALT1 axis with MALT1 inhibitors may become an effective strategy to treat DVT by suppressing thromboinflammation without inducing bleeding complications.


Subject(s)
Thrombosis , Venous Thrombosis , Adaptor Proteins, Signal Transducing , Animals , B-Cell CLL-Lymphoma 10 Protein/metabolism , CARD Signaling Adaptor Proteins/metabolism , Endothelial Cells/metabolism , Guanylate Kinases/metabolism , Inflammasomes/metabolism , Inflammation , Inflammation Mediators , Interleukin-1beta/metabolism , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , NF-kappa B/metabolism , P-Selectin/metabolism , Thromboinflammation , Thromboplastin/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Venous Thrombosis/genetics , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...