Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.056
Filter
2.
Commun Biol ; 7(1): 944, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39098885

ABSTRACT

Depression is a prevalent and intricate mental disorder. The involvement of small RNA molecules, such as microRNAs in the pathogenesis and neuronal mechanisms underlying the depression have been documented. Previous studies have demonstrated the involvement of microRNA-143-3p (miR-143-3p) in the process of fear memory and pathogenesis of ischemia; however, the relationship between miR-143-3p and depression remains poorly understood. Here we utilized two kinds of mouse models to investigate the role of miR-143-3p in the pathogenesis of depression. Our findings reveal that the expression of miR-143-3p is upregulated in the ventral hippocampus (VH) of mice subjected to chronic restraint stress (CRS) or acute Lipopolysaccharide (LPS) treatment. Inhibiting the expression of miR-143-3p in the VH effectively alleviates depressive-like behaviors in CRS and LPS-treated mice. Furthermore, we identify Lasp1 as one of the downstream target genes regulated by miR-143-3p. The miR-143-3p/Lasp1 axis primarily affects the occurrence of depressive-like behaviors in mice by modulating synapse numbers in the VH. Finally, miR-143-3p/Lasp1-induced F-actin change is responsible for the synaptic number variations in the VH. In conclusion, this study enhances our understanding of microRNA-mediated depression pathogenesis and provides novel prospects for developing therapeutic approaches for this intractable mood disorder.


Subject(s)
Cytoskeletal Proteins , Depression , Hippocampus , MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Hippocampus/metabolism , Mice , Depression/metabolism , Depression/genetics , Male , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Mice, Inbred C57BL , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Behavior, Animal , Disease Models, Animal , Stress, Psychological/metabolism , Gene Expression Regulation
3.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125895

ABSTRACT

The branched architecture of neuronal dendrites is a key factor in how neurons form ordered networks and discoveries continue to be made identifying proteins and protein-protein interactions that direct or execute the branching and extension of dendrites. Our prior work showed that the molecular scaffold Pdlim5 and delta-catenin, in conjunction, are two proteins that help regulate the branching and elongation of dendrites in cultured hippocampal neurons and do so through a phosphorylation-dependent mechanism triggered by upstream glutamate signaling. In this report we have focused on Pdlim5's multiple scaffolding domains and how each contributes to dendrite branching. The three identified regions within Pdlim5 are the PDZ, DUF, and a trio of LIM domains; however, unresolved is the intra-molecular conformation of Pdlim5 as well as which domains are essential to regulate dendritic branching. We address Pdlim5's structure and function by examining the role of each of the domains individually and using deletion mutants in the context of the full-length protein. Results using primary hippocampal neurons reveal that the Pdlim5 DUF domain plays a dominant role in increasing dendritic branching. Neither the PDZ domain nor the LIM domains alone support increased branching. The central role of the DUF domain was confirmed using deletion mutants in the context of full-length Pdlim5. Guided by molecular modeling, additional domain mapping studies showed that the C-terminal LIM domain forms a stable interaction with the N-terminal PDZ domain, and we identified key amino acid residues at the interface of each domain that are needed for this interaction. We posit that the central DUF domain of Pdlim5 may be subject to modulation in the context of the full-length protein by the intra-molecular interaction between the N-terminal PDZ and C-terminal LIM domains. Overall, our studies reveal a novel mechanism for the regulation of Pdlim5's function in the regulation of neuronal branching and highlight the critical role of the DUF domain in mediating these effects.


Subject(s)
Dendrites , Hippocampus , LIM Domain Proteins , PDZ Domains , Dendrites/metabolism , Animals , Hippocampus/metabolism , Hippocampus/cytology , LIM Domain Proteins/metabolism , LIM Domain Proteins/chemistry , LIM Domain Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Protein Domains , Neurons/metabolism , Rats , Cells, Cultured , Humans
4.
Nat Immunol ; 25(9): 1623-1636, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39107403

ABSTRACT

Targeting tumor-infiltrating regulatory T (TI-Treg) cells is a potential strategy for cancer therapy. The ATPase p97 in complex with cofactors (such as Npl4) has been investigated as an antitumor drug target; however, it is unclear whether p97 has a function in immune cells or immunotherapy. Here we show that thonzonium bromide is an inhibitor of the interaction of p97 and Npl4 and that this p97-Npl4 complex has a critical function in TI-Treg cells. Thonzonium bromide boosts antitumor immunity without affecting peripheral Treg cell homeostasis. The p97-Npl4 complex bridges Stat3 with E3 ligases PDLIM2 and PDLIM5, thereby promoting Stat3 degradation and enabling TI-Treg cell development. Collectively, this work shows an important role for the p97-Npl4 complex in controlling Treg-TH17 cell balance in tumors and identifies possible targets for immunotherapy.


Subject(s)
T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , Animals , Mice , Humans , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Nuclear Proteins/metabolism , Neoplasms/immunology , Cell Line, Tumor , Th17 Cells/immunology , Immunotherapy/methods , LIM Domain Proteins/metabolism , Adenosine Triphosphatases/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Female
5.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(4): 541-552, 2024 Apr 28.
Article in English, Chinese | MEDLINE | ID: mdl-39019783

ABSTRACT

OBJECTIVES: Super-enhancer-associated genes may be closely related to the progression of osteosarcoma, curcumin exhibits a certain inhibitory effect on tumors such as osteosarcoma. This study aims to investigate the effects of curcumin on osteosarcoma in vitro and in vivo, and to determine whether curcumin can inhibit the progression of osteosarcoma by suppressing the expression of super-enhancer-associated genes LIM and senescent cell antigen-like-containing domain 1 (LIMS1), secreted protein acidic and rich in cysteine (SPARC), and sterile alpha motif domain containing 4A (SAMD4A). METHODS: Human osteosarcoma cell lines (MG63 cells or U2OS cells) were treated with 5 to 50 µmol/L curcumin for 24, 48, and 72 hours, followed by the methyl thiazolyl tetrazolium (MTT) assay to detect cell viability. Cells were incubated with dimethyl sulfoxide (DMSO) or curcumin (2.5, 5.0 µmol/L) for 7 days, and a colony formation assay was used to measure in vitro cell proliferation. After treatment with DMSO or curcumin (10, 15 µmol/L), a scratch healing assay and a transwell migration assay were performed to evaluate cell migration ability. Real-time reverse transcription polymerase chain reaction (real-time RT-PCR) and Western blotting were used to detect mRNA and protein expression levels of LIMS1, SPARC, and SAMD4A in the cells. An osteosarcoma-bearing nude mouse model was established, and curcumin was administered via gavage for 14 days to assess the impact of curcumin on tumor volume and weight in vivo. Real-time RT-PCR was used to measure mRNA expression levels of LIMS1, SPARC, and SAMD4A in the cancer and adjacent tissues from 12 osteosarcoma patients. RESULTS: After treating cells with different concentrations of curcumin for 24, 48, and 72 hours, cell viability were all significantly decreased. Compared with the DMSO group, the colony formation rates in the 2.5 µmol/L and 5.0 µmol/L curcumin groups significantly declined (both P<0.01). The scratch healing assay showed that, compared with the DMSO group, the migration rates of cells in the 10 µmol/L and 15 µmol/L curcumin groups were significantly reduced. The exception was the 10 µmol/L curcumin group at 24 h, where the migration rate of U2OS cells did not show a statistically significant difference (P>0.05), while all other differences were statistically significant (P<0.01 or P<0.001). The transwell migration assay results showed that the number of migrating cells in the 10 µmol/L and 15 µmol/L curcumin groups was significantly lower than that in the DMSO group (both P<0.001). In the in vivo tumor-bearing mouse experiment, the curcumin group showed a reduction in tumor mass (P<0.01) and a significant reduction in tumor volume (P<0.001) compared with the control group. Compared with the DMSO group, the mRNA expression levels of LIMS1, SPARC, and SAMD4A in the 10 µmol/L and 15 µmol/L curcumin groups were significantly down-regulated (all P<0.05). Additionally, the protein expression level of LIMS1 in U2OS cells in the 10 µmol/L curcumin group was significantly lower than that in the DMSO group (P<0.05). Compared with adjacent tissues, the mRNA expression level of SPARC in osteosarcoma tissues was significantly increased (P<0.001), while the mRNA expression levels of LIMS1 and SAMD4A did not show statistically significant differences (both P>0.05). CONCLUSIONS: Curcumin inhibits the proliferation and migration of osteosarcoma both in vitro and in vivo, which may be associated with the inactivation of super-enhancer-associated gene LIMS1.


Subject(s)
Bone Neoplasms , Cell Movement , Cell Proliferation , Curcumin , Mice, Nude , Osteonectin , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Curcumin/pharmacology , Humans , Cell Proliferation/drug effects , Cell Movement/drug effects , Animals , Bone Neoplasms/genetics , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Cell Line, Tumor , Mice , Osteonectin/genetics , Osteonectin/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C
6.
Oncol Rep ; 52(3)2024 09.
Article in English | MEDLINE | ID: mdl-38994754

ABSTRACT

Cancer constitutes a multifaceted ailment characterized by the dysregulation of numerous genes and pathways. Among these, LIM domain only 7 (LMO7) has emerged as a significant player in various cancer types, garnering substantial attention for its involvement in tumorigenesis and cancer progression. This review endeavors to furnish a comprehensive discourse on the functional intricacies and mechanisms of LMO7 in cancer, with a particular emphasis on its potential as both a therapeutic target and prognostic indicator. It delves into the molecular attributes of LMO7, its implications in cancer etiology and the underlying mechanisms propelling its oncogenic properties. Furthermore, it underscores the extant challenges and forthcoming prospects in targeting LMO7 for combating cancer.


Subject(s)
LIM Domain Proteins , Neoplasms , Transcription Factors , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Prognosis , Carcinogenesis/genetics , Molecular Targeted Therapy/methods
7.
Cell Adh Migr ; 18(1): 4-17, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39076043

ABSTRACT

Lim Domain and Actin Binding protein1 (lima1) influence cancer cell function. Thus far, functional role of lima1 in cholangiocarcinoma remains unknown. We used public databases, in vitro experiments, and multi-omics analysis to investigate the Lima1 in cholangiocarcinoma. Our results showed that lima1 expression is significantly upregulated and high levels of lima1 are significantly associated with vascular invasion in cholangiocarcinoma. Furthermore, lima1 knocking out inhibits the RBE cell invasion. Multi-omics data suggest that lima1 affect a broad spectrum of cancer related pathways, promoting tumor progression and metastatic ability in cholangiocarcinoma. This study provides insights into molecular associations of lima1 with tumorigenesist and establishes a preliminary picture of the correlation network in cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Gene Expression Regulation, Neoplastic , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Cholangiocarcinoma/genetics , Humans , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/genetics , Cell Line, Tumor , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Cell Movement/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Cell Proliferation , Neoplasm Invasiveness , Male , Female
8.
Cell Rep ; 43(7): 114480, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39003737

ABSTRACT

The cytoskeleton of the cell is constantly exposed to physical forces that regulate cellular functions. Selected members of the LIM (Lin-11, Isl-1, and Mec-3) domain-containing protein family accumulate along force-bearing actin fibers, with evidence supporting that the LIM domain is solely responsible for this force-induced interaction. However, LIM domain's force-induced interactions are not limited to actin. LIMK1 and LMO1, both containing only two tandem LIM domains, are recruited to force-bearing keratin fibers in epithelial cells. This unique recruitment is mediated by their LIM domains and regulated by the sequences outside the LIM domains. Based on in vitro reconstitution of this interaction, LIMK1 and LMO1 directly interact with stretched keratin 8/18 fibers. These results show that LIM domain's mechano-sensing abilities extend to the keratin cytoskeleton, highlighting the diverse role of LIM proteins in force-regulated signaling.


Subject(s)
Intermediate Filaments , Keratins , LIM Domain Proteins , Lim Kinases , LIM Domain Proteins/metabolism , Humans , Lim Kinases/metabolism , Keratins/metabolism , Intermediate Filaments/metabolism , Protein Binding , Animals , Transcription Factors/metabolism
9.
Dev Biol ; 515: 46-58, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38968989

ABSTRACT

The Daam1 protein regulates Wnt-induced cytoskeletal changes during vertebrate gastrulation though its full mode of action and binding partners remain unresolved. Here we identify Reversion Induced LIM domain protein (RIL) as a new interacting protein of Daam1. Interaction studies uncover binding of RIL to the C-terminal actin-nucleating portion of Daam1 in a Wnt-responsive manner. Immunofluorescence studies showed subcellular localization of RIL to actin fibers and co-localization with Daam1 at the plasma membrane. RIL gain- and loss-of-function approaches in Xenopus produced severe gastrulation defects in injected embryos. Additionally, a simultaneous loss of Daam1 and RIL synergized to produce severe gastrulation defects indicating RIL and Daam1 may function in the same signaling pathway. RIL further synergizes with another novel Daam1-interacting protein, Formin Binding Protein 1 (FNBP1), to regulate gastrulation. Our studies altogether show RIL mediates Daam1-regulated non-canonical Wnt signaling that is required for vertebrate gastrulation.


Subject(s)
Actin Cytoskeleton , Gastrulation , Microfilament Proteins , Wnt Signaling Pathway , Xenopus Proteins , Xenopus laevis , Animals , Female , Humans , Rats , Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Protein Binding , Wnt Signaling Pathway/physiology , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics
10.
Dev Biol ; 515: 67-78, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38968988

ABSTRACT

Sequence-specific transcription factors often function as components of large regulatory complexes. LIM-domain binding protein (LDB) and single-stranded DNA-binding protein (SSDP) function as core scaffolds of transcriptional complexes in animals and plants. Little is known about potential partners and functions for LDB/SSDP complexes in the context of tissue regeneration. In this work, we find that planarian LDB1 and SSDP2 promote tissue regeneration, with a particular function in anterior regeneration and mediolateral polarity reestablishment. We find that LDB1 and SSDP2 interact with one another and with characterized planarian LIM-HD proteins Arrowhead, Islet1, and Lhx1/5-1. We also show that SSDP2 and LDB1 function with islet1 in polarity reestablishment and with lhx1/5-1 in serotonergic neuron maturation. Finally, we find new roles for LDB1 and SSDP2 in regulating gene expression in the planarian intestine and parenchyma; these functions are likely LIM-HD-independent. Together, our work provides insight into LDB/SSDP complexes in a highly regenerative organism. Further, our work provides a strong starting point for identifying and characterizing potential binding partners of LDB1 and SSDP2 and for exploring roles for these proteins in diverse aspects of planarian physiology.


Subject(s)
Body Patterning , Planarians , Regeneration , Transcription Factors , Animals , Planarians/genetics , Planarians/physiology , Regeneration/genetics , Regeneration/physiology , Transcription Factors/metabolism , Transcription Factors/genetics , Body Patterning/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Helminth Proteins/genetics , Helminth Proteins/metabolism , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Gene Expression Regulation, Developmental
11.
Mol Cancer ; 23(1): 143, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992675

ABSTRACT

BACKGROUND: Emerging evidence indicates the pivotal involvement of circular RNAs (circRNAs) in cancer initiation and progression. Understanding the functions and underlying mechanisms of circRNAs in tumor development holds promise for uncovering novel diagnostic indicators and therapeutic targets. In this study, our focus was to elucidate the function and regulatory mechanism of hsa-circ-0003764 in hepatocellular carcinoma (HCC). METHODS: A newly discovered hsa-circ-0003764 (circPTPN12) was identified from the circbase database. QRT-PCR analysis was utilized to assess the expression levels of hsa-circ-0003764 in both HCC tissues and cells. We conducted in vitro and in vivo experiments to examine the impact of circPTPN12 on the proliferation and apoptosis of HCC cells. Additionally, RNA-sequencing, RNA immunoprecipitation, biotin-coupled probe pull-down assays, and FISH were employed to confirm and establish the relationship between hsa-circ-0003764, PDLIM2, OTUD6B, P65, and ESRP1. RESULTS: In HCC, the downregulation of circPTPN12 was associated with an unfavorable prognosis. CircPTPN12 exhibited suppressive effects on the proliferation of HCC cells both in vitro and in vivo. Mechanistically, RNA sequencing assays unveiled the NF-κB signaling pathway as a targeted pathway of circPTPN12. Functionally, circPTPN12 was found to interact with the PDZ domain of PDLIM2, facilitating the ubiquitination of P65. Furthermore, circPTPN12 bolstered the assembly of the PDLIM2/OTUD6B complex by promoting the deubiquitination of PDLIM2. ESRP1 was identified to bind to pre-PTPN12, thereby fostering the generation of circPTPN12. CONCLUSIONS: Collectively, our findings indicate the involvement of circPTPN12 in modulating PDLIM2 function, influencing HCC progression. The identified ESRP1/circPTPN12/PDLIM2/NF-κB axis shows promise as a novel therapeutic target in the context of HCC.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Gene Expression Regulation, Neoplastic , LIM Domain Proteins , Liver Neoplasms , NF-kappa B , RNA, Circular , RNA-Binding Proteins , Signal Transduction , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , RNA, Circular/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , NF-kappa B/metabolism , Mice , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line, Tumor , Disease Progression , Apoptosis/genetics , Prognosis , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Male , Female , Mice, Nude
12.
Eur J Med Res ; 29(1): 309, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38831471

ABSTRACT

The long non-coding RNA (lncRNA) Small Nucleolar RNA Host Gene 4 (SNHG4) has been demonstrated to be significantly downregulated in various inflammatory conditions, yet its role in chronic obstructive pulmonary disease (COPD) remains elusive. This study aims to elucidate the biological function of SNHG4 in COPD and to unveil its potential molecular targets. Our findings reveal that both SNHG4 and Four and a Half LIM Domains 1 (FHL1) were markedly downregulated in COPD, whereas microRNA-409-3p (miR-409-3p) was upregulated. Importantly, SNHG4 exhibited a negative correlation with inflammatory markers in patients with COPD, but a positive correlation with forced expiratory volume in 1s percentage (FEV1%). SNHG4 distinguished COPD patients from non-smokers with high sensitivity, specificity, and accuracy. Overexpression of SNHG4 ameliorated cigarette smoke extract (CSE)-mediated inflammation, apoptosis, oxidative stress, and airway remodeling in 16HBE bronchial epithelial cells. These beneficial effects of SNHG4 overexpression were reversed by the overexpression of miR-409-3p or the silencing of FHL1. Mechanistically, SNHG4 competitively bound to miR-409-3p, mediating the expression of FHL1, and consequently improving inflammation, apoptosis, oxidative stress, and airway remodeling in 16HBE cells. Additionally, SNHG4 regulated the miR-409-3p/FHL1 axis to inhibit the activation of the mitogen-activated protein kinase (MAPK) pathway induced by CSE. In a murine model of COPD, knockdown of SNHG4 exacerbated CSE-induced pulmonary inflammation, apoptosis, and oxidative stress. In summary, our data affirm that SNHG4 mitigates pulmonary inflammation, apoptosis, and oxidative damage mediated by COPD through the regulation of the miR-409-3p/FHL1 axis.


Subject(s)
Airway Remodeling , Apoptosis , Cell Proliferation , MicroRNAs , Pulmonary Disease, Chronic Obstructive , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis/genetics , Airway Remodeling/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Cell Proliferation/genetics , Animals , Mice , Male , MAP Kinase Signaling System/genetics , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Inflammation/metabolism , Inflammation/genetics , Female , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Middle Aged , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice, Inbred C57BL
13.
J Exp Clin Cancer Res ; 43(1): 169, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38880883

ABSTRACT

BACKGROUND: Cancer is characterized by dysregulated cellular metabolism. Thus, understanding the mechanisms underlying these metabolic alterations is important for developing targeted therapies. In this study, we investigated the pro-tumoral effect of PDZ and LIM domain 2 (PDLIM2) downregulation in lung cancer growth and its association with the accumulation of mitochondrial ROS, oncometabolites and the activation of hypoxia-inducible factor-1 (HIF-1) α in the process. METHODS: Databases and human cancer tissue samples were analyzed to investigate the roles of PDLIM2 and HIF-1α in cancer growth. DNA microarray and gene ontology enrichment analyses were performed to determine the cellular functions of PDLIM2. Seahorse assay, flow cytometric analysis, and confocal microscopic analysis were employed to study mitochondrial functions. Oncometabolites were analyzed using liquid chromatography-mass spectrometry (LC-MS). A Lewis lung carcinoma (LLC) mouse model was established to assess the in vivo function of PDLIM2 and HIF-1α. RESULTS: The expression of PDLIM2 was downregulated in lung cancer, and this downregulation correlated with poor prognosis in patients. PDLIM2 highly regulated genes associated with mitochondrial functions. Mechanistically, PDLIM2 downregulation resulted in NF-κB activation, impaired expression of tricarboxylic acid (TCA) cycle genes particularly the succinate dehydrogenase (SDH) genes, and mitochondrial dysfunction. This disturbance contributed to the accumulation of succinate and other oncometabolites, as well as the buildup of mitochondrial reactive oxygen species (mtROS), leading to the activation of hypoxia-inducible factor 1α (HIF-1α). Furthermore, the expression of HIF-1α was increased in all stages of lung cancer. The expression of PDLIM2 and HIF-1α was reversely correlated in lung cancer patients. In the animal study, the orally administered HIF-1α inhibitor, PX-478, significantly reduces PDLIM2 knockdown-promoted tumor growth. CONCLUSION: These findings shed light on the complex action of PDLIM2 on mitochondria and HIF-1α activities in lung cancer, emphasizing the role of HIF-1α in the tumor-promoting effect of PDLIM2 downregulation. Additionally, they provide new insights into a strategy for precise targeted treatment by suggesting that HIF-1α inhibitors may serve as therapy for lung cancer patients with PDLIM2 downregulation.


Subject(s)
Down-Regulation , Hypoxia-Inducible Factor 1, alpha Subunit , LIM Domain Proteins , Mitochondria , Reactive Oxygen Species , Humans , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Cell Line, Tumor , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/genetics , Gene Expression Regulation, Neoplastic , Female , Male
14.
Proc Natl Acad Sci U S A ; 121(24): e2320867121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838015

ABSTRACT

O-GlcNAcase (OGA) is the only human enzyme that catalyzes the hydrolysis (deglycosylation) of O-linked beta-N-acetylglucosaminylation (O-GlcNAcylation) from numerous protein substrates. OGA has broad implications in many challenging diseases including cancer. However, its role in cell malignancy remains mostly unclear. Here, we report that a cancer-derived point mutation on the OGA's noncatalytic stalk domain aberrantly modulates OGA interactome and substrate deglycosylation toward a specific set of proteins. Interestingly, our quantitative proteomic studies uncovered that the OGA stalk domain mutant preferentially deglycosylated protein substrates with +2 proline in the sequence relative to the O-GlcNAcylation site. One of the most dysregulated substrates is PDZ and LIM domain protein 7 (PDLIM7), which is associated with the tumor suppressor p53. We found that the aberrantly deglycosylated PDLIM7 suppressed p53 gene expression and accelerated p53 protein degradation by promoting the complex formation with E3 ubiquitin ligase MDM2. Moreover, deglycosylated PDLIM7 significantly up-regulated the actin-rich membrane protrusions on the cell surface, augmenting the cancer cell motility and aggressiveness. These findings revealed an important but previously unappreciated role of OGA's stalk domain in protein substrate recognition and functional modulation during malignant cell progression.


Subject(s)
Cytoskeleton , LIM Domain Proteins , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Cytoskeleton/metabolism , Acetylglucosamine/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Cell Line, Tumor , Glycosylation , Hydrolysis , Mutation , Cell Movement , Antigens, Neoplasm , Hyaluronoglucosaminidase , Histone Acetyltransferases
15.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928252

ABSTRACT

Inclusion body myositis (IBM) is a slowly progressive disorder belonging to the idiopathic inflammatory myopathies, and it represents the most common adult-onset acquired myopathy. The main clinical features include proximal or distal muscular asymmetric weakness, with major involvement of long finger flexors and knee extensors. The main histological findings are the presence of fiber infiltrations, rimmed vacuoles, and amyloid inclusions. The etiopathogenesis is a challenge because both environmental and genetic factors are implicated in muscle degeneration and a distinction has been made previously between sporadic and hereditary forms. Here, we describe an Italian patient affected with a hereditary form of IBM with onset in his mid-forties. Next-generation sequencing analysis disclosed a heterozygous mutation c.76C>T (p.Pro26Ser) in the PDZ motif of the LDB3/ZASP gene, a mutation already described in a family with a late-onset myopathy and highly heterogenous degree of skeletal muscle weakness. In the proband's muscle biopsy, the expression of ZASP, myotilin, and desmin were increased. In our family, in addition to the earlier age of onset, the clinical picture is even more peculiar given the evidence, in one of the affected family members, of complete ophthalmoplegia in the vertical gaze. These findings help extend our knowledge of the clinical and genetic background associated with inclusion body myopathic disorders.


Subject(s)
LIM Domain Proteins , Myositis, Inclusion Body , Pedigree , Humans , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/pathology , Male , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Middle Aged , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Mutation , Adult
16.
BMC Oral Health ; 24(1): 729, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918827

ABSTRACT

BACKGROUND: Despite the better prognosis associated with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC), some patients experience relapse and succumb to the disease; thus, there is a need for biomarkers identifying these patients for intensified treatment. Leucine-rich repeats and immunoglobulin-like domain (LRIG) protein 1 is a negative regulator of receptor tyrosine kinase signaling and a positive prognostic factor in OPSCC. Studies indicate that LRIG1 interacts with the LIM domain 7 protein (LMO7), a stabilizer of adherence junctions. Its role in OPSCC has not been studied before. METHODS: A total of 145 patients diagnosed with OPSCC were enrolled. Immunohistochemical LMO7 expression and staining intensity were evaluated in the tumors and correlated with known clinical and pathological prognostic factors, such as HPV status and LRIG1, CD44, Ki67, and p53 expression. RESULTS: Our results show that high LMO7 expression is associated with significantly longer overall survival (OS) (p = 0.044). LMO7 was a positive prognostic factor for OS in univariate analysis (HR 0.515, 95% CI: 0.267-0.994, p = 0.048) but not in multivariate analysis. The LMO7 expression correlated with LRIG1 expression (p = 0.048), consistent with previous findings. Interestingly, strong LRIG1 staining intensity was an independent negative prognostic factor in the HPV-driven group of tumors (HR 2.847, 95% Cl: 1.036-7.825, p = 0.043). CONCLUSIONS: We show for the first time that high LMO7 expression is a positive prognostic factor in OPSCC, and we propose that LMO7 should be further explored as a biomarker. In contrast to previous reports, LRIG1 expression was shown to be an independent negative prognostic factor in HPV-driven OPSCC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Squamous Cell , LIM Domain Proteins , Oropharyngeal Neoplasms , Humans , Oropharyngeal Neoplasms/virology , Oropharyngeal Neoplasms/metabolism , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/mortality , Male , Female , Middle Aged , Prognosis , LIM Domain Proteins/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Aged , Transcription Factors/metabolism , Membrane Glycoproteins/metabolism , Adult , Ki-67 Antigen/metabolism , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/analysis , Tumor Suppressor Protein p53/metabolism , Papillomavirus Infections/complications , Immunohistochemistry , Aged, 80 and over , Survival Rate
17.
Arch Dermatol Res ; 316(7): 401, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878083

ABSTRACT

BACKGROUND: The adhesive properties of vitiligo melanocytes have decreased under oxidative stress., cytoskeleton proteins can control cell adhesion. Paeoniflorin (PF) was proved to resist hydrogen peroxide (H2O2)-induced oxidative stress in melanocytes via nuclear factorE2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. OBJECTIVES: This study was to investigate whether PF exerts anti-oxidative effect through influencing cytoskeleton markers or potential signaling pathway. METHODS: Human Oxidative Stress Plus array was used to identify the differentially expressed genes between H2O2 + PF group and H2O2 only group, in PIG1 and PIG3V melanocyte cell lines respectively. Western blotting was used to verify the PCR array results and to test the protein expression levels of cytoskeleton markers including Ras homolog family member A (RhoA), Rho-associated kinase 1 (ROCK1) and antioxidative marker Nrf2. Small interfering RNA was used to knock down PDZ and LIM domain 1 (PDLIM1). RESULTS: PF increased the expressions of PDLIM1, RhoA and ROCK1 in H2O2-induced PIG1, in contrast, decreased the expressions of PDLIM1 and ROCK1 in H2O2-induced PIG3V. Knockdown of PDLIM1 increased the expressions of RhoA and Nrf2 in PF-pretreated H2O2-induced PIG1, and ROCK1 and Nrf2 in PF-pretreated H2O2-induced PIG3V. CONCLUSIONS: PF regulates RhoA/ROCK1 and Nrf2 pathways in PDLIM1-dependent or independent manners in H2O2-induced melanocytes. In PIG1, PF promotes PDLIM1 to inhibit RhoA/ROCK1 pathway or activates Nrf2/HO-1 pathway, separately. In PIG3V, PF directly downregulates ROCK1 in PDLIM1-independent manner or upregulates Nrf2 dependent of PDLIM1.


Subject(s)
Glucosides , Hydrogen Peroxide , LIM Domain Proteins , Melanocytes , Monoterpenes , NF-E2-Related Factor 2 , Oxidative Stress , Signal Transduction , rho-Associated Kinases , rhoA GTP-Binding Protein , NF-E2-Related Factor 2/metabolism , rho-Associated Kinases/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Humans , Glucosides/pharmacology , Oxidative Stress/drug effects , rhoA GTP-Binding Protein/metabolism , Hydrogen Peroxide/metabolism , Signal Transduction/drug effects , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Monoterpenes/pharmacology , Cell Line
18.
Biochem Biophys Res Commun ; 727: 150277, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38936225

ABSTRACT

With the aging of the global demographic, the prevention and treatment of osteoporosis are becoming crucial issues. The gradual loss of self-renewal and osteogenic differentiation capabilities in bone marrow stromal cells (BMSCs) is one of the key factors contributing to osteoporosis. To explore the regulatory mechanisms of BMSCs differentiation, we collected bone marrow cells of femoral heads from patients undergoing total hip arthroplasty for single-cell RNA sequencing analysis. Single-cell RNA sequencing revealed significantly reduced CRIP1 (Cysteine-Rich Intestinal Protein 1) expression and osteogenic capacity in the BMSCs of osteoporosis patients compared to non-osteoporosis group. CRIP1 is a gene that encodes a member of the LIM/double zinc finger protein family, which is involved in the regulation of various cellular processes including cell growth, development, and differentiation. CRIP1 knockdown resulted in decreased alkaline phosphatase activity, mineralization and expression of osteogenic markers, indicating impaired osteogenic differentiation. Conversely, CRIP1 overexpression, both in vitro and in vivo, enhanced osteogenic differentiation and rescued bone mass reduction in ovariectomy-induced osteoporosis mice model. The study further established CRIP1's modulation of osteogenesis through the Wnt signaling pathway, suggesting that targeting CRIP1 could offer a novel approach for osteoporosis treatment by promoting bone formation and preventing bone loss.


Subject(s)
Cell Differentiation , LIM Domain Proteins , Mesenchymal Stem Cells , Osteoblasts , Osteogenesis , Osteoporosis , Wnt Signaling Pathway , Osteogenesis/genetics , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Differentiation/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Humans , Osteoblasts/metabolism , Osteoblasts/cytology , Female , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Mice , Mice, Inbred C57BL , Cells, Cultured , Middle Aged , Aged , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Carrier Proteins
19.
Nat Commun ; 15(1): 4945, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858386

ABSTRACT

Single administration of low-dose ketamine has both acute and sustained anti-depressant effects. Sustained effect is associated with restoration of glutamatergic synapses in medial prefrontal cortic (mFPC) neurons. Ketamine induced profound changes in a number of molecular pathways in a mouse model for chronic stress. Cell-cell communication analyses predicted that planar-cell-polarity (PCP) signaling was decreased after chronic administration of corticosterone but increased following ketamine administration in most of the excitatory neurons. Similar decrease of PCP signaling in excitatory neurons was predicted in dorsolateral prefrontal cortical (dl-PFC) neurons of patients with major depressive disorder (MDD). We showed that the basolateral amygdala (BLA)-projecting infralimbic prefrontal cortex (IL PFC) neurons regulate immobility time in the tail suspension test and food consumption. Conditionally knocking out Celsr2 and Celsr3 or Prickle2 in the BLA-projecting IL PFC neurons abolished ketamine-induced synapse restoration and behavioral remission. Therefore, PCP proteins in IL PFC-BLA neurons mediate synapse restoration induced by of low-dose ketamine.


Subject(s)
Disease Models, Animal , Ketamine , Neurons , Prefrontal Cortex , Synapses , Animals , Ketamine/pharmacology , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Synapses/drug effects , Synapses/metabolism , Neurons/metabolism , Neurons/drug effects , Mice , Male , Humans , Cell Polarity/drug effects , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/drug therapy , Mice, Knockout , Stress, Psychological , Corticosterone , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/drug effects , Mice, Inbred C57BL , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Glutamic Acid/metabolism , Antidepressive Agents/pharmacology
20.
J Neurophysiol ; 132(2): 418-432, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38838299

ABSTRACT

The appropriate growth of the neurons, accurate organization of their synapses, and successful neurotransmission are indispensable for sensorimotor activities. These processes are highly dynamic and tightly regulated. Extensive genetic, molecular, physiological, and behavioral studies have identified many molecular candidates and investigated their roles in various neuromuscular processes. In this article, we show that Beadex (Bx), the Drosophila LIM only (LMO) protein, is required for motor activities and neuromuscular growth of Drosophila. The larvae bearing Bx7, a null allele of Bx, and the RNAi-mediated neuronal-specific knockdown of Bx show drastically reduced crawling behavior, a diminished synaptic span of the neuromuscular junctions (NMJs) and an increased spontaneous neuronal firing with altered motor patterns in the central pattern generators (CPGs). Microarray studies identified multiple targets of Beadex that are involved in different cellular and molecular pathways, including those associated with the cytoskeleton and mitochondria that could be responsible for the observed neuromuscular defects. With genetic interaction studies, we further show that Highwire (Hiw), a negative regulator of synaptic growth at the NMJs, negatively regulates Bx, as the latter's deficiency was able to rescue the phenotype of the Hiw null mutant, HiwDN. Thus, our data indicate that Beadex functions downstream of Hiw to regulate the larval synaptic growth and physiology.NEW & NOTEWORTHY A novel role for Beadex (Bx) regulates the larval neuromuscular junction (NMJ) structure and function in a tissue-specific manner. Bx is expressed in a subset of Toll-6-expressing neurons and is involved in regulating synaptic span and physiology, possibly through its negative interaction with Highwire (Hiw). The findings of this study provide insights into the molecular mechanisms underlying NMJ development and function and warrant further investigation to understand the role of Bx in these processes fully.


Subject(s)
Drosophila Proteins , Larva , Neuromuscular Junction , Animals , Neuromuscular Junction/physiology , Neuromuscular Junction/metabolism , Neuromuscular Junction/growth & development , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Larva/growth & development , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Drosophila melanogaster/growth & development , Central Pattern Generators/physiology , Central Pattern Generators/metabolism , Drosophila
SELECTION OF CITATIONS
SEARCH DETAIL