Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.816
Filter
1.
Biosci Rep ; 44(5)2024 May 29.
Article in English | MEDLINE | ID: mdl-38808367

ABSTRACT

Platelets are small anucleate blood cells supporting vascular function. They circulate in a quiescent state monitoring the vasculature for injuries. Platelets adhere to injury sites and can be rapidly activated to secrete granules and to form platelet/platelet aggregates. These responses are controlled by signalling networks that include G proteins and their regulatory guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Recent proteomics studies have revealed the complete spectrum of G proteins, GEFs, and GAPs present in platelets. Some of these proteins are specific for platelets and very few have been characterised in detail. GEFs and GAPs play a major role in setting local levels of active GTP-bound G proteins in response to activating and inhibitory signals encountered by platelets. Thus, GEFs and GAPs are highly regulated themselves and appear to integrate G protein regulation with other cellular processes. This review focuses on GAPs of small G proteins of the Arf, Rab, Ras, and Rho families, as well as of heterotrimeric G proteins found in platelets.


Subject(s)
Blood Platelets , GTPase-Activating Proteins , Blood Platelets/metabolism , Humans , GTPase-Activating Proteins/metabolism , Animals , GTP-Binding Proteins/metabolism , Signal Transduction , Guanine Nucleotide Exchange Factors/metabolism
2.
Cancer Biol Ther ; 25(1): 2343450, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38742566

ABSTRACT

The potential function and mechanism of circRNAs in regulating malignant performances of Osteosarcoma (OS) cells have not been well investigated. The expression level of CircLMO7, miR-21-5p and ARHGAP24 were detected by RT-qPCR. The relationship between miR-21-5p and circ-LMO7, as well as between miR-21-5p and ARHGAP24, was predicted and examined through bioinformatics analysis and luciferase reporter gene experiments. Moreover, OS cell growth, invasion, migration, and apoptosis were detected using the cell counting kit-8 (CCK-8), transwell and flow cytometry assays, respectively. ARHGAP24 protein level was measured using western blotting. In present study, we choose to investigate the role and mechanism of circ-LOM7 on OS cell proliferation, migration and invasion. circ-LOM7 was found to be down-regulated in OS tissues and cell lines. Enforced expression of circ-LOM7 suppressed the growth, invasion, and migration of OS cells. In contrast, decreasing circ-LMO7 expression had opposite effects. Furthermore, miR-21-5p was predicted to be sponged by circ-LMO7, and had an opposite role of circ-LMO7 in OS. Moreover, ARHGAP24 served as miR-21-5p's downstream target. Mechanistically, circ-LMO7 was packed in exosomes and acted as a cancer-suppresser on OS by sponging miR-21-5p and upregulating the expression of ARHGAP24. The exosomal circ-LMO7 expression was significantly decreased in OS cell exosomes, and co-culture experiments showed that exosomal circ-LMO7 suppressed the proliferation ability of OS cells. Circ-LMO7 exerts as a tumor suppressor in OS, and the circ-LMO7/miR-21-5P/ARHGAP24 axis is involved in OS progression.


Subject(s)
Disease Progression , Exosomes , GTPase-Activating Proteins , MicroRNAs , Osteosarcoma , RNA, Circular , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Exosomes/metabolism , Exosomes/genetics , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Cell Proliferation , Mice , Animals , Cell Line, Tumor , Cell Movement/genetics , Apoptosis/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Male , Female
3.
Sci Rep ; 14(1): 11790, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38783033

ABSTRACT

The Rho GTPase activating protein family (ARHGAPs) is expressed in pancreatic adenocarcinoma (PAAD) but its function is unclear. The aim of this study was to explore the role and potential clinical value of ARHGAPs in PAAD. Using TCGA and GEO databases to analyze expression of ARHGAPs in PAAD and normal tissues. Survival curve was drawn by Kaplan-Meier. ARHGAPs were integrated analyzed by GEPIA2, TIMER, UCLCAN, cBioPortal and R language. Protein level and prognostic value were evaluated via IHC staining or survival analysis. We totally identify 18 differentially expressed (DE) ARHGAPs in PAAD. Among the 18 DE genes, 8 were positively correlated with tumor grade; abnorrmal expression of 5 was positively correlated with copy number variation; expression of 4 was positively correlated with promoter hypomethylation. Multivariate Cox regression identified ARHGAP5, ARHGAP11A, and ARHGAP12 as independent prognostic factors of PAAD. The function of ARHGAPs was mainly related to GTPase activity and signaling, axon guidance, proteoglycans in cancer and focal adhesion. Expression of 7 ARHGAPs was strongly correlated with immune infiltration. Immunohistochemistry showed increased protein levels of ARHGAP5, ARHGAP11A, and ARHGAP12 in PAAD tissues. Survival analysis confirmed a negative correlation between ARHGAP5, ARHGAP11A, and ARHGAP12 expression and patient prognosis. Multivariate Cox regression proved ARHGAP5, ARHGAP11A, and ARHGAP12 could serve as independent prognostic indicators for PAAD. Finally, this study verified ARHGAP5, ARHGAP11A, and ARHGAP12 as independent prognostic factors in PAAD, suggesting their significance for the diagnosis and treatment of PAAD.


Subject(s)
Adenocarcinoma , GTPase-Activating Proteins , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms , Humans , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/mortality , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/mortality , Prognosis , Male , Female , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Aged , DNA Methylation , Kaplan-Meier Estimate , DNA Copy Number Variations
4.
Genes (Basel) ; 15(5)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38790165

ABSTRACT

Adams-Oliver syndrome is a rare inherited condition characterized by scalp defects and limb abnormalities. It is caused by variants in different genes such as ARHGAP31. Here, we used an interdisciplinary approach to study a family with lower limb anomalies. We identified a novel variant in the ARHGAP31 gene that is predicted to result in a truncated protein with a constitutively activated catalytic site due to the loss of 688 amino acids involved in the C-terminal domain, essential for protein auto-inhibition. Pathogenic variants in ARHGAP31 exon 12, leading to a premature protein termination, are associated with Adams-Oliver syndrome. Bioinformatic analysis was useful to elucidate the impact of the identified genetic variant on protein structure. To better understand the impact of the identified variant, 3D protein models were predicted for the ARHGAP31 wild type, the newly discovered variant, and other pathogenetic alterations already reported. Our study identified a novel variant probably involved in Adams-Oliver syndrome and increased the evidence on the phenotypic variability in patients affected by this syndrome, underlining the importance of translational research, including experimental and bioinformatics analyses. This strategy represents a successful model to investigate molecular mechanisms involved in syndrome occurrence.


Subject(s)
Ectodermal Dysplasia , GTPase-Activating Proteins , Pedigree , Phenotype , Scalp Dermatoses , Humans , GTPase-Activating Proteins/genetics , Scalp Dermatoses/genetics , Scalp Dermatoses/congenital , Scalp Dermatoses/pathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Male , Female , Mutation , Limb Deformities, Congenital/genetics , Phosphoproteins
5.
Commun Biol ; 7(1): 602, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762624

ABSTRACT

The role of endothelial cells in promoting cancer cell extravasation to the brain during the interaction of cancer cells with the vasculature is not well characterised. We show that brain endothelial cells activate EGFR signalling in triple-negative breast cancer cells with propensity to metastasise to the brain. This activation is dependent on soluble factors secreted by brain endothelial cells, and occurs via the RAC1 GEF DOCK4, which is required for breast cancer cell extravasation to the brain in vivo. Knockdown of DOCK4 inhibits breast cancer cell entrance to the brain without affecting cancer cell survival or growth. Defective extravasation is associated with loss of elongated morphology preceding intercalation into brain endothelium. We also show that brain endothelial cells promote paracrine stimulation of mesenchymal-like morphology of breast cancer cells via DOCK4, DOCK9, RAC1 and CDC42. This stimulation is accompanied by EGFR activation necessary for brain metastatic breast cancer cell elongation which can be reversed by the EGFR inhibitor Afatinib. Our findings suggest that brain endothelial cells promote metastasis through activation of cell signalling that renders breast cancer cells competent for extravasation. This represents a paradigm of brain endothelial cells influencing the signalling and metastatic competency of breast cancer cells.


Subject(s)
Brain Neoplasms , Brain , Endothelial Cells , ErbB Receptors , Signal Transduction , rac1 GTP-Binding Protein , ErbB Receptors/metabolism , ErbB Receptors/genetics , Humans , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Female , Endothelial Cells/metabolism , Endothelial Cells/pathology , Cell Line, Tumor , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Mice , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics
6.
Commun Biol ; 7(1): 596, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762629

ABSTRACT

Apicomplexan parasites harbor a complex endomembrane system as well as unique secretory organelles. These complex cellular structures require an elaborate vesicle trafficking system, which includes Rab GTPases and their regulators, to assure the biogenesis and secretory of the organelles. Here we exploit the model apicomplexan organism Toxoplasma gondii that encodes a family of Rab GTPase Activating Proteins, TBC (Tre-2/Bub2/Cdc16) domain-containing proteins. Functional profiling of these proteins in tachyzoites reveals that TBC9 is the only essential regulator, which is localized to the endoplasmic reticulum (ER) in T. gondii strains. Detailed analyses demonstrate that TBC9 is required for normal distribution of proteins targeting to the ER, and the Golgi apparatus in the parasite, as well as for the normal formation of daughter inner membrane complexes (IMCs). Pull-down assays show a strong protein interaction between TBC9 and specific Rab GTPases (Rab11A, Rab11B, and Rab2), supporting the role of TBC9 in daughter IMC formation and early vesicular transport. Thus, this study identifies the only essential TBC domain-containing protein TBC9 that regulates early vesicular transport and IMC formation in T. gondii and potentially in closely related protists.


Subject(s)
Endoplasmic Reticulum , GTPase-Activating Proteins , Protozoan Proteins , Toxoplasma , rab GTP-Binding Proteins , Toxoplasma/metabolism , Toxoplasma/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Endoplasmic Reticulum/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Golgi Apparatus/metabolism , Protein Transport , Animals , Transport Vesicles/metabolism
7.
Sci Rep ; 14(1): 12153, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802496

ABSTRACT

Hypoxia-inducible factors (HIF) 1 and 2 regulate similar but distinct sets of target genes. Although HIFs are best known for their roles in mediating the hypoxia response accumulating evidence suggests that under certain conditions HIFs, particularly HIF2, may function also under normoxic conditions. Here we report that HIF2α functions under normoxic conditions in kidney epithelial cells to regulate formation of adherens junctions. HIF2α expression was required to induce Dock4/Rac1/Pak1-signaling mediating stability and compaction of E-cadherin at nascent adherens junctions. Impaired adherens junction formation in HIF2α- or Dock4-deficient cells led to aberrant cyst morphogenesis in 3D kidney epithelial cell cultures. Taken together, we show that HIF2α functions in normoxia to regulate epithelial morphogenesis.


Subject(s)
Adherens Junctions , Basic Helix-Loop-Helix Transcription Factors , Cell Polarity , Signal Transduction , rac1 GTP-Binding Protein , Adherens Junctions/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , rac1 GTP-Binding Protein/metabolism , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Cadherins/metabolism , Cadherins/genetics , Mice , Humans , Epithelial Cells/metabolism , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Cell Line
8.
PeerJ ; 12: e17362, 2024.
Article in English | MEDLINE | ID: mdl-38766486

ABSTRACT

Backgrounds: TBC1D family members (TBC1Ds) are a group of proteins that contain the Tre2-Bub2-Cdc16 (TBC) domain. Recent studies have shown that TBC1Ds are involved in tumor growth, but no analysis has been done of expression patterns and prognostic values of TBC1Ds in hepatocellular carcinoma (HCC). Methods: The expression levels of TBC1Ds were evaluated in HCC using the TIMER, UALCN and Protein Atlas databases. The correlation between the mRNA levels of TBC1Ds and the prognosis of patients with HCC in the GEPIA database was then analyzed. An enrichment analysis then revealed genes that potentially interact with TBC1Ds. The correlation between levels of TBC1Ds and tumor-infiltrating immune cells (TIICs) in HCC were studied using the TIMER 2.0 database. Finally, a series of in vitro assays verified the role of TBC1Ds in HCC progression. Results: This study revealed the upregulated expression of TBC1Ds in HCC and the strong positive correlation between the mRNA levels of TBC1Ds and poor prognosis of patients with HCC. The functions of TBC1Ds were mainly related to autophagy and the AMPK pathway. There was also a significant correlation between level of TBC1Ds and tumor-infiltrating immune cells (TIICs) in HCC. The promoting role of TBC1Ds in HCC progression was verified in vitro assays. Conclusion: The results of this analysis indicate that TBC1Ds may serve as new biomarkers for early diagnosis and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , GTPase-Activating Proteins , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Prognosis , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Autophagy/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Line, Tumor
9.
J Cell Sci ; 137(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38563084

ABSTRACT

Angiogenesis is a tightly controlled dynamic process demanding a delicate equilibrium between pro-angiogenic signals and factors that promote vascular stability. The spatiotemporal activation of the transcriptional co-factors YAP (herein referring to YAP1) and TAZ (also known WWTR1), collectively denoted YAP/TAZ, is crucial to allow for efficient collective endothelial migration in angiogenesis. The focal adhesion protein deleted-in-liver-cancer-1 (DLC1) was recently described as a transcriptional downstream target of YAP/TAZ in endothelial cells. In this study, we uncover a negative feedback loop between DLC1 expression and YAP activity during collective migration and sprouting angiogenesis. In particular, our study demonstrates that signaling via the RhoGAP domain of DLC1 reduces nuclear localization of YAP and its transcriptional activity. Moreover, the RhoGAP activity of DLC1 is essential for YAP-mediated cellular processes, including the regulation of focal adhesion turnover, traction forces, and sprouting angiogenesis. We show that DLC1 restricts intracellular cytoskeletal tension by inhibiting Rho signaling at the basal adhesion plane, consequently reducing nuclear YAP localization. Collectively, these findings underscore the significance of DLC1 expression levels and its function in mitigating intracellular tension as a pivotal mechanotransductive feedback mechanism that finely tunes YAP activity throughout the process of sprouting angiogenesis.


Subject(s)
Focal Adhesions , GTPase-Activating Proteins , Mechanotransduction, Cellular , Tumor Suppressor Proteins , YAP-Signaling Proteins , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Movement , Feedback, Physiological , Focal Adhesions/metabolism , Focal Adhesions/genetics , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Mechanotransduction, Cellular/genetics , Neovascularization, Physiologic , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins/metabolism
10.
Curr Biol ; 34(10): 2049-2065.e6, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38677281

ABSTRACT

Plants rely on autophagy and membrane trafficking to tolerate stress, combat infections, and maintain cellular homeostasis. However, the molecular interplay between autophagy and membrane trafficking is poorly understood. Using an AI-assisted approach, we identified Rab3GAP-like (Rab3GAPL) as a key membrane trafficking node that controls plant autophagy negatively. Rab3GAPL suppresses autophagy by binding to ATG8, the core autophagy adaptor, and deactivating Rab8a, a small GTPase essential for autophagosome formation and defense-related secretion. Rab3GAPL reduces autophagic flux in three model plant species, suggesting that its negative regulatory role in autophagy is conserved in land plants. Beyond autophagy regulation, Rab3GAPL modulates focal immunity against the oomycete pathogen Phytophthora infestans by preventing defense-related secretion. Altogether, our results suggest that Rab3GAPL acts as a molecular rheostat to coordinate autophagic flux and defense-related secretion by restraining Rab8a-mediated trafficking. This unprecedented interplay between a RabGAP-Rab pair and ATG8 sheds new light on the intricate membrane transport mechanisms underlying plant autophagy and immunity.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Autophagy , GTPase-Activating Proteins , Plant Immunity , Autophagy/physiology , Arabidopsis/immunology , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis/microbiology , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Protein 8 Family/genetics , Phytophthora infestans/physiology , Plant Diseases/microbiology , Plant Diseases/immunology , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Protein Transport
11.
Curr Biol ; 34(10): 2132-2146.e5, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38688282

ABSTRACT

Actin cortex patterning and dynamics are critical for cell shape changes. These dynamics undergo transitions during development, often accompanying changes in collective cell behavior. Although mechanisms have been established for individual cells' dynamic behaviors, the mechanisms and specific molecules that result in developmental transitions in vivo are still poorly understood. Here, we took advantage of two developmental systems in Drosophila melanogaster to identify conditions that altered cortical patterning and dynamics. We identified a Rho guanine nucleotide exchange factor (RhoGEF) and Rho GTPase activating protein (RhoGAP) pair required for actomyosin waves in egg chambers. Specifically, depletion of the RhoGEF, Ect2, or the RhoGAP, RhoGAP15B, disrupted actomyosin wave induction, and both proteins relocalized from the nucleus to the cortex preceding wave formation. Furthermore, we found that overexpression of a different RhoGEF and RhoGAP pair, RhoGEF2 and Cumberland GAP (C-GAP), resulted in actomyosin waves in the early embryo, during which RhoA activation precedes actomyosin assembly by ∼4 s. We found that C-GAP was recruited to actomyosin waves, and disrupting F-actin polymerization altered the spatial organization of both RhoA signaling and the cytoskeleton in waves. In addition, disrupting F-actin dynamics increased wave period and width, consistent with a possible role for F-actin in promoting delayed negative feedback. Overall, we showed a mechanism involved in inducing actomyosin waves that is essential for oocyte development and is general to other cell types, such as epithelial and syncytial cells.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , GTPase-Activating Proteins , Animals , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Actomyosin/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Female , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Embryo, Nonmammalian/metabolism , Body Patterning
13.
Int J Mol Sci ; 25(8)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38674130

ABSTRACT

IQGAP3 (IQ Motif Containing GTPase Activating Protein 3) is member of the IQGAP family of scaffold proteins, which are essential for assembling multiprotein complexes that coordinate various intracellular signaling pathways. Previous research has shown that IQGAP3 is overexpressed in psoriatic skin lesions. Given its involvement in processes like cell proliferation and chemokine signaling, we sought to explore its molecular role in driving the psoriatic phenotype of keratinocytes. By conducting transcriptome profiling of HaCaT keratinocytes, we identified numerous psoriasis-associated pathways that were affected when IQGAP3 was knocked down. These included alterations in NFkB signaling, EGFR signaling, activation of p38/MAPK and ERK1/ERK2, lipid metabolism, cytokine production, and the response to inflammatory cytokine stimulation. Real-time analysis further revealed changes in cell growth dynamics, including proliferation and wound healing. The balance between cell proliferation and apoptosis was altered, as were skin barrier functions and the production of IL-6 and IFNγ. Despite these significant findings, the diversity of the alterations observed in the knockdown cells led us to conclude that IQGAP3 may not be the best target for the therapeutic inhibition to normalize the phenotype of keratinocytes in psoriasis.


Subject(s)
Cell Proliferation , GTPase-Activating Proteins , Keratinocytes , Psoriasis , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Psoriasis/metabolism , Psoriasis/pathology , Psoriasis/genetics , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Signal Transduction , HaCaT Cells , Cytokines/metabolism , Apoptosis , Skin/metabolism , Skin/pathology , Cell Line , Gene Expression Profiling
14.
Medicine (Baltimore) ; 103(16): e37702, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38640279

ABSTRACT

RATIONALE: Hereditary hearing loss is known to exhibit a significant degree of genetic heterogeneity. Herein, we present a case report of a novel mutation in the tenascin-C (TNC) gene in Chinese patients with nonsyndromic hearing loss (NSHL). PATIENT CONCERNS: This includes a young deaf couple and their 2-year-old baby. DIAGNOSES: Based on the clinical information, hearing test, metagenomic next-generation sequencing (mNGS), Sanger sequencing, protein function and structure analysis, and model prediction, in our case, the study results revealed 2 heterozygous mutations in the TNC gene (c.2852C>T, p.Thr951Ile) and the TBC1 domain family member 24 (TBC1D24) gene (c.1570C>T, p.Arg524Trp). These mutations may be responsible for the hearing loss observed in this family. Notably, the heterozygous mutations in the TNC gene (c.2852C>T, p.Thr951Ile) have not been previously reported in the literature. INTERVENTIONS: Avoid taking drugs that can cause deafness, wearing hearing AIDS, and cochlear implants. OUTCOMES: Regular follow-up of family members is ongoing. LESSONS: The genetic diagnosis of NSHL holds significant importance as it helps in making informed treatment decisions, providing prognostic information, and offering genetic counseling for the patient's family.


Subject(s)
Deafness , Hearing Loss, Sensorineural , Hearing Loss , Tenascin , Child, Preschool , Humans , China , Deafness/genetics , GTPase-Activating Proteins/genetics , Hearing Loss/genetics , Hearing Loss, Sensorineural/genetics , Mutation , Pedigree , Tenascin/genetics
15.
Am J Physiol Endocrinol Metab ; 326(6): E807-E818, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38656130

ABSTRACT

One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle from rodents and humans of both sexes. We recently found that concurrent mutation of three key sites to prevent their phosphorylation (Ser588, Thr642, and Ser704) on Akt substrate of 160 kDa (AS160; also known as TBC1D4) reduced the magnitude of the enhancement of postexercise ISGU (PEX-ISGU) by muscle from male, but not female rats. However, we did not test the role of individual phosphorylation sites on PEX-ISGU. Accordingly, our current aim was to test whether AS160 Ser704 phosphorylation (pSer704) is required for elevated PEX-ISGU by muscle. AS160-knockout (AS160-KO) rats (female and male) were studied when either in sedentary or 3 h after acute exercise. Adeno-associated virus (AAV) vectors were used to enable muscle expression of wild-type AS160 (AAV-WT-AS160) or AS160 mutated Ser704 to alanine to prevent phosphorylation (AAV-1P-AS160). Paired epitrochlearis muscles from each rat were injected with AAV-WT-AS160 or AAV-1P-AS160. We discovered that regardless of sex 1) AS160 abundance in AS160-KO rats was similar in paired muscles expressing WT-AS160 versus 1P-AS160; 2) muscles from exercised versus sedentary rats had greater ISGU, and PEX-ISGU was slightly greater for muscles expressing 1P-AS160 versus contralateral muscles expressing WT-AS160; and 3) pAS160Thr642 was lower in muscles expressing 1P-AS160 versus paired muscles expressing WT-AS160. These results indicate that pAS160Ser704 was not essential for elevated PEX-ISGU by skeletal muscle from rats of either sex. Furthermore, elimination of the postexercise increase in pAS160Thr642 did not lessen the postexercise effect on ISGU.NEW & NOTEWORTHY The current study evaluated the role of Akt substrate of 160 kDa (AS160) phosphorylation on Ser704 in increased insulin-stimulated glucose uptake by skeletal muscle after exercise. Adeno-associated virus vectors were engineered to express either wild-type-AS160 or AS160 mutated so that it could not be phosphorylated on Ser704 in paired muscles from AS160-knockout rats. The results demonstrated that AS160 phosphorylation on Ser704 was not essential for exercise-induced elevation in insulin-stimulated glucose uptake by rats of either sex.


Subject(s)
GTPase-Activating Proteins , Glucose , Insulin , Muscle, Skeletal , Physical Conditioning, Animal , Animals , Female , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Rats , Phosphorylation , Physical Conditioning, Animal/physiology , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Insulin/metabolism , Glucose/metabolism , Serine/metabolism , Rats, Sprague-Dawley
16.
Nat Commun ; 15(1): 3468, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658571

ABSTRACT

Metabolism has recently emerged as a major target of genes implicated in the evolutionary expansion of human neocortex. One such gene is the human-specific gene ARHGAP11B. During human neocortex development, ARHGAP11B increases the abundance of basal radial glia, key progenitors for neocortex expansion, by stimulating glutaminolysis (glutamine-to-glutamate-to-alpha-ketoglutarate) in mitochondria. Here we show that the ape-specific protein GLUD2 (glutamate dehydrogenase 2), which also operates in mitochondria and converts glutamate-to-αKG, enhances ARHGAP11B's ability to increase basal radial glia abundance. ARHGAP11B + GLUD2 double-transgenic bRG show increased production of aspartate, a metabolite essential for cell proliferation, from glutamate via alpha-ketoglutarate and the TCA cycle. Hence, during human evolution, a human-specific gene exploited the existence of another gene that emerged during ape evolution, to increase, via concerted changes in metabolism, progenitor abundance and neocortex size.


Subject(s)
GTPase-Activating Proteins , Glutamate Dehydrogenase , Neocortex , Neocortex/metabolism , Neocortex/embryology , Neocortex/growth & development , Neocortex/cytology , Humans , Animals , Glutamate Dehydrogenase/metabolism , Glutamate Dehydrogenase/genetics , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Ketoglutaric Acids/metabolism , Neuroglia/metabolism , Glutamic Acid/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Mice , Citric Acid Cycle/genetics , Female
17.
Int Immunopharmacol ; 133: 112050, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38636370

ABSTRACT

Thyroid cancer (THCA) is the most common endocrine malignancy worldwide and has been rising at the fastest rate in recent years. Long-stranded non-coding RNAs (lncRNAs) and N6-methyladenosine (m6A) have been associated with immunotherapy efficacy and cancer prognosis. However, how m6A-associated lncRNAs (mrlncRNAs) affect the prognosis of patients with thyroid cancer is unclear. Therefore, this study utilized The Cancer Genome Atlas (TCGA) database to provide thyroid cancer-related transcriptomic data and related clinical data. The R program was used to identify m6A-related lncRNAs, and a risk model consisting of two lncRNAs (LINC02471 and DOCK9-DT) was obtained using least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Kaplan-Meier survival analysis and transient subject operating characteristics (ROC) were used for analysis. The results showed a substantial association between immune cell infiltration and risk scores. Independent analyses confirmed that the expression of LINC02471 and DOCK9-DT was significantly higher in thyroid cancer tissues than in normal tissues, suggesting that they may be useful biomarkers for thyroid cancer.


Subject(s)
Adenosine , Biomarkers, Tumor , RNA, Long Noncoding , Thyroid Neoplasms , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Biomarkers, Tumor/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/immunology , Adenosine/analogs & derivatives , Adenosine/metabolism , Gene Expression Regulation, Neoplastic , Prognosis , Male , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Middle Aged
18.
Biol Open ; 13(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682696

ABSTRACT

Arf GTPase-activating proteins (ArfGAPs) mediate the hydrolysis of GTP bound to ADP-ribosylation factors. ArfGAPs are critical for cargo sorting in the Golgi-to-ER traffic. However, the role of ArfGAPs in sorting into intralumenal vesicles (ILVs) in multivesicular bodies (MVBs) in post-Golgi traffic remains unclear. Exosomes are extracellular vesicles (EVs) of endosomal origin. CD63 is an EV marker. CD63 is enriched ILVs in MVBs of cells. However, the secretion of CD63 positive EVs has not been consistent with the data on CD63 localization in MVBs, and how CD63-containing EVs are formed is yet to be understood. To elucidate the mechanism of CD63 transport to ILVs, we focused on CD63 localization in MVBs and searched for the ArfGAPs involved in CD63 localization. We observed that ADAP1 and ARAP1 depletion inhibited CD63 localization to enlarged endosomes after Rab5Q79L overexpression. We tested epidermal growth factor (EGF) and CD9 localization in MVBs. We observed that ADAP1 and ARAP1 depletion inhibited CD9 localization in enlarged endosomes but not EGF. Our results indicate ADAP1 and ARAP1, regulate incorporation of CD63 and CD9, but not EGF, in overlapped and different MVBs. Our work will contribute to distinguish heterogenous ILVs and exosomes by ArfGAPs.


Subject(s)
Adaptor Proteins, Signal Transducing , GTPase-Activating Proteins , Multivesicular Bodies , Tetraspanin 30 , Tetraspanin 30/metabolism , Humans , Multivesicular Bodies/metabolism , GTPase-Activating Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Protein Transport , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Endosomes/metabolism , HeLa Cells , Carrier Proteins
19.
Oncol Res ; 32(4): 659-678, 2024.
Article in English | MEDLINE | ID: mdl-38560572

ABSTRACT

Background: IQGAP3 plays a crucial role in regulating cell proliferation, division, and cytoskeletal organization. Abnormal expression of IQGAP3 has been linked to various tumors, but its function in glioma is not well understood. Methods: Various methods, including genetic differential analysis, single-cell analysis, ROC curve analysis, Cox regression, Kaplan-Meier analysis, and enrichment analysis, were employed to analyze the expression patterns, diagnostic potential, prognostic implications, and biological processes involving IQGAP3 in normal and tumor tissues. The impact of IQGAP3 on immune infiltration and the immune microenvironment in gliomas was evaluated using immunofluorescence. Additionally, the cBioPortal database was used to analyze copy number variations and mutation sites of IQGAP3. Experimental validation was also performed to assess the effects of IQGAP3 on glioma cells and explore underlying mechanisms. Results: High IQGAP3 expression in gliomas is associated with an unfavorable prognosis, particularly in wild-type IDH and 1p/19q non-codeleted gliomas. Enrichment analysis revealed that IQGAP3 is involved in regulating the cell cycle, PI3K/AKT signaling, p53 signaling, and PLK1-related pathways. Furthermore, IQGAP3 expression may be closely related to the immunosuppressive microenvironment of glioblastoma. BRD-K88742110 and LY-303511 are potential drugs for targeting IQGAP3 in anti-glioma therapy. In vitro experiments showed that downregulation of IQGAP3 inhibits the proliferation and migration of glioma cells, with the PLK1/PI3K/AKT pathway potentially playing a crucial role in IQGAP3-mediated glioma progression. Conclusion: IQGAP3 shows promise as a valuable biomarker for diagnosis, prognosis, and immunotherapeutic strategies in gliomas.


Subject(s)
Brain Neoplasms , Glioma , Humans , Prognosis , Brain Neoplasms/pathology , DNA Copy Number Variations , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Glioma/pathology , Tumor Microenvironment/genetics , GTPase-Activating Proteins
20.
PLoS One ; 19(4): e0294227, 2024.
Article in English | MEDLINE | ID: mdl-38564630

ABSTRACT

Current evidence suggests that DEP domain containing 1 (DEPDC1) has an important effect on non-small-cell lung cancer (NSCLC). However, the diagnostic value and the regulatory function within NSCLC are largely unclear. This work utilized publicly available databases and in vitro experiments for exploring, DEPDC1 expression, clinical features, diagnostic significance and latent molecular mechanism within NSCLC. According to our results, DEPDC1 was remarkably upregulated in the tissues of NSCLC patients compared with non-carcinoma tissues, linked with gender, stage, T classification and N classification based on TCGA data and associated with smoking status and stage according to GEO datasets. Meanwhile, the summary receiver operating characteristic (sROC) curve analysis result showed that DEPDC1 had a high diagnostic value in NSCLC (AUC = 0.96, 95% CI: 0.94-0.98; diagnostic odds ratio = 99.08, 95%CI: 31.91-307.65; sensitivity = 0.89, 95%CI: 0.81-0.94; specificity = 0.92, 95%CI: 0.86-0.96; positive predictive value = 0.94, 95%CI: 0.89-0.98; negative predictive value = 0.78, 95%CI: 0.67-0.90; positive likelihood ratio = 11.77, 95%CI: 6.11-22.68; and negative likelihood ratio = 0.12, 95%CI: 0.06-0.22). Subsequently, quantitative real-time PCR (qRT-PCR) and western blotting indicated that DEPDC1 was high expressed in NSCLC cells. According to the in vitro MTS and apoptotic assays, downregulated DEPDC1 expression targeting P53 signaling pathway inhibited the proliferation of NSCLC cells while promoting apoptosis of NSCLC cells. Moreover, DEPDC1 was significantly correlated with immune cell infiltrating levels in NSCLC based on TCGA data, which were primarily associated with T cells CD4 memory activated, macrophages M1, B cells memory, mast cells resting, T cells regulatory, monocytes, and T cells CD4 memory resting. Compared with the group with high expression of DEPDC1, the group with low expression level had higher scores for immune checkpoint inhibitors (ICIs) treatment. GSEA confirmed that DEPDC1 was involved in gene expression and tumor-related signaling pathways. Finally, DEPDC1 and its associated immune-related genes were shown to be enriched in 'receptor ligand activity', 'external side of plasma membrane', 'regulation of innate immune response', and 'Epstein-Barr virus infection' pathways. The present study demonstrates that DEPDC1 may contribute to NSCLC tumorigenesis and can be applied as the biomarker for diagnosis and immunology.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Epstein-Barr Virus Infections , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Herpesvirus 4, Human/metabolism , Signal Transduction , Neoplasm Proteins/genetics , GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...