Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.691
Filter
1.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000312

ABSTRACT

Hepatocellular carcinoma (HCC) is a common tumor. Our group has previously reported that sorcin (SRI) plays an important role in the progression and prognosis of HCC. This study aims to explore the mechanism of SRI inhibiting the mitochondrial apoptosis. Bioinformatics analysis, co-IP and immunofluorescence were used to analyze the relationship between SRI and STAT3. MMP and Hoechst staining were performed to detect the effect of SRI on cell apoptosis. The expression of apoptosis-related proteins and NF-κB signaling pathway were examined by Western blot and immunohistochemistry when SRI overexpression or underexpression in vivo and in vitro were found. Moreover, inhibitors were used to further explore the molecular mechanism. Overexpression of SRI inhibited cell apoptosis, which was attenuated by SRI knockdown in vitro and in vivo. Moreover, we identified that STAT3 is an SRI-interacting protein. Mechanistically, SRI interacts with STAT3 and then activates the NF-κB signaling pathway in vitro and in vivo. SRI interacting with STAT3 inhibits apoptosis by the NF-κB pathway and further contributes to the proliferation in HCC, which offers a novel clue and a new potential therapeutic target for HCC.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Liver Neoplasms , Mitochondria , NF-kappa B , STAT3 Transcription Factor , Signal Transduction , STAT3 Transcription Factor/metabolism , Humans , Apoptosis/drug effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Animals , Mice , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cell Proliferation/drug effects , Cell Line, Tumor , Mice, Nude , Male , Gene Expression Regulation, Neoplastic/drug effects
2.
Nat Commun ; 15(1): 5564, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956119

ABSTRACT

Chemical probes are an indispensable tool for translating biological discoveries into new therapies, though are increasingly difficult to identify since novel therapeutic targets are often hard-to-drug proteins. We introduce FRASE-based hit-finding robot (FRASE-bot), to expedite drug discovery for unconventional therapeutic targets. FRASE-bot mines available 3D structures of ligand-protein complexes to create a database of FRAgments in Structural Environments (FRASE). The FRASE database can be screened to identify structural environments similar to those in the target protein and seed the target structure with relevant ligand fragments. A neural network model is used to retain fragments with the highest likelihood of being native binders. The seeded fragments then inform ultra-large-scale virtual screening of commercially available compounds. We apply FRASE-bot to identify ligands for Calcium and Integrin Binding protein 1 (CIB1), a promising drug target implicated in triple negative breast cancer. FRASE-based virtual screening identifies a small-molecule CIB1 ligand (with binding confirmed in a TR-FRET assay) showing specific cell-killing activity in CIB1-dependent cancer cells, but not in CIB1-depletion-insensitive cells.


Subject(s)
Antineoplastic Agents , Calcium-Binding Proteins , Drug Discovery , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Ligands , Drug Discovery/methods , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/chemistry , Cell Line, Tumor , Computer Simulation , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Protein Binding , Neural Networks, Computer
3.
Methods Mol Biol ; 2842: 167-178, 2024.
Article in English | MEDLINE | ID: mdl-39012595

ABSTRACT

In this chapter, we present an experimental protocol to conduct DNA methylation editing experiments, that is, to induce loss or gain of DNA methylation, targeting Dlk1-Dio3 imprinted domain, a well-studied imprinted locus, in ES cells. In this protocol, plasmid vectors expressing the DNA methylation editing tools, combining the CRISPR/dCas9 system and the SunTag system coupled to a DNA methyltransferase or a TET enzyme, are introduced into cells for transient expression. By employing this strategy, researchers can effectively investigate a distinct DNA methylation signature that has an impact on the imprinting status, including gene expression and histone modifications, across the entire domain. We also describe strategies for allele-specific quantitative analyses of DNA methylation, gene expression, and histone modifications and binding protein levels for assessing the imprinting state of the locus.


Subject(s)
CRISPR-Cas Systems , DNA Methylation , Gene Editing , Genomic Imprinting , Gene Editing/methods , Animals , Mice , Genetic Loci , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Iodide Peroxidase/genetics , Alleles , Humans
4.
Nat Commun ; 15(1): 5567, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956087

ABSTRACT

Diabetes involves the death or dysfunction of pancreatic ß-cells. Analysis of bulk sequencing from human samples and studies using in vitro and in vivo models suggest that endoplasmic reticulum and inflammatory signaling play an important role in diabetes progression. To better characterize cell type-specific stress response, we perform multiplexed single-cell RNA sequencing to define the transcriptional signature of primary human islet cells exposed to endoplasmic reticulum and inflammatory stress. Through comprehensive pair-wise analysis of stress responses across pancreatic endocrine and exocrine cell types, we define changes in gene expression for each cell type under different diabetes-associated stressors. We find that ß-, α-, and ductal cells have the greatest transcriptional response. We utilize stem cell-derived islets to study islet health through the candidate gene CIB1, which was upregulated under stress in primary human islets. Our findings provide insights into cell type-specific responses to diabetes-associated stress and establish a resource to identify targets for diabetes therapeutics.


Subject(s)
Endoplasmic Reticulum Stress , Insulin-Secreting Cells , Islets of Langerhans , Humans , Endoplasmic Reticulum Stress/genetics , Islets of Langerhans/metabolism , Insulin-Secreting Cells/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Single-Cell Analysis , Glucagon-Secreting Cells/metabolism , Sequence Analysis, RNA , Transcriptome , Stress, Physiological
5.
Sci Rep ; 14(1): 15091, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956220

ABSTRACT

Fibulin-2 is a multidomain, disulfide-rich, homodimeric protein which belongs to a broader extracellular matrix family. It plays an important role in the development of elastic fiber structures. Malfunction of fibulin due to mutation or poor expression can result in a variety of diseases including synpolydactyly, limb abnormalities, eye disorders leading to blindness, cardiovascular diseases and cancer. Traditionally, fibulins have either been produced in mammalian cell systems or were isolated from the extracellular matrix, a procedure that results in poor availability for structural and functional studies. Here, we produced seven fibulin-2 constructs covering 62% of the mature protein (749 out of 1195 residues) using a prokaryotic expression system. Biophysical studies confirm that the purified constructs are folded and that the presence of disulfide bonds within the constructs makes them extremely thermostable. In addition, we solved the first crystal structure for any fibulin isoform, a structure corresponding to the previously suggested three motifs related to anaphylatoxin. The structure reveals that the three anaphylatoxins moieties form a single-domain structure.


Subject(s)
Calcium-Binding Proteins , Humans , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Protein Stability , Protein Domains
6.
Sci Adv ; 10(27): eadl1197, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959305

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by increasing fibrosis, which can enhance tumor progression and spread. Here, we undertook an unbiased temporal assessment of the matrisome of the highly metastatic KPC (Pdx1-Cre, LSL-KrasG12D/+, LSL-Trp53R172H/+) and poorly metastatic KPflC (Pdx1-Cre, LSL-KrasG12D/+, Trp53fl/+) genetically engineered mouse models of pancreatic cancer using mass spectrometry proteomics. Our assessment at early-, mid-, and late-stage disease reveals an increased abundance of nidogen-2 (NID2) in the KPC model compared to KPflC, with further validation showing that NID2 is primarily expressed by cancer-associated fibroblasts (CAFs). Using biomechanical assessments, second harmonic generation imaging, and birefringence analysis, we show that NID2 reduction by CRISPR interference (CRISPRi) in CAFs reduces stiffness and matrix remodeling in three-dimensional models, leading to impaired cancer cell invasion. Intravital imaging revealed improved vascular patency in live NID2-depleted tumors, with enhanced response to gemcitabine/Abraxane. In orthotopic models, NID2 CRISPRi tumors had less liver metastasis and increased survival, highlighting NID2 as a potential PDAC cotarget.


Subject(s)
Carcinoma, Pancreatic Ductal , Fibrosis , Pancreatic Neoplasms , Proteomics , Animals , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Proteomics/methods , Mice , Humans , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Disease Models, Animal , Cell Line, Tumor , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Gemcitabine , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Cell Adhesion Molecules
7.
J Mol Model ; 30(8): 248, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38965105

ABSTRACT

CONTEXT: Calcium-dependent signaling in plants is responsible for several major cellular events, including the activation of the salinity-responsive pathways. Calcium binds to calcineurin B-like protein (CBL), and the resulting CBL-Ca2+ complex binds to CBL-interacting protein kinase (CIPK). The CBL-CIPK complex enhances the CIPK interaction with an upstream kinase. The upstream kinase phosphorylates CIPK that, in turn, phosphorylates membrane transporters. Phosphorylation influences transporter activity to kick-start many downstream functions, such as balancing the cytosolic Na+-to-K+ ratio. The CBL-CIPK interaction is pivotal for Ca2+-dependent salinity stress signaling. METHODS: Computational methods are used to model the entire Arabidopsis thaliana CIPK24 protein structure in its autoinhibited and open-activated states. Arabidopsis thaliana CIPK24-CBL4 complex is predicted based on the protein-protein docking methods. The available structural and functional data support the CIPK24 and the CIPK24-CBL4 complex models. Models are energy-minimized and subjected to molecular dynamics (MD) simulations. MD simulations for 500 ns and 300 ns enabled us to predict the importance of conserved residues of the proteins. Finally, the work is extended to predict the CIPK24-CBL4 complex with the upstream kinase GRIK2. MD simulation for 300 ns on the ternary complex structure enabled us to identify the critical CIPK24-GRIK2 interactions. Together, these data could be used to engineer the CBL-CIPK interaction network for developing salt tolerance in crops.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Calcium-Binding Proteins , Molecular Dynamics Simulation , Protein Serine-Threonine Kinases , Salt Stress , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry , Arabidopsis/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/chemistry , Protein Binding , Phosphorylation , Molecular Docking Simulation
8.
Respir Res ; 25(1): 267, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970088

ABSTRACT

BACKGROUND: Lung cancer is the second most common cancer with the highest mortality in the world. Calumenin as a molecular chaperone that not only binds various proteins within the endoplasmic reticulum but also plays crucial roles in diverse processes associated with tumor development. However, the regulatory mechanism of calumenin in lung adenocarcinoma remains elusive. Here, we studied the impact of calumenin on lung adenocarcinoma and explored possible mechanisms. METHODS: 5-ethynyl-2'-deoxyuridine assay, colony formation, transwell and wound healing assays were performed to explore the effects of calumenin on the proliferation and migration of lung adenocarcinoma cells. To gain insights into the underlying mechanisms through which calumenin knockdown inhibits the migration and proliferation of lung adenocarcinoma, we performed Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, Gene Set Enrichment Analysis and Ingenuity Pathway Analysis based on transcriptomics by comparing calumenin knockdown with normal A549 cells. RESULTS: The mRNA and protein levels of calumenin in lung adenocarcinoma are highly expressed and they are related to an unfavorable prognosis in this disease. Calumenin enhances the proliferation and migration of A549 and H1299 cells. Gene Set Enrichment Analysis revealed that knockdown of calumenin in A549 cells significantly inhibited MYC and V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog signaling pathways while activating interferon signals, inflammatory signals, and p53 pathways. Ingenuity pathway analysis provided additional insights, indicating that the interferon and inflammatory pathways were prominently activated upon calumenin knockdown in A549 cells. CONCLUSIONS: The anti-cancer mechanism of calumenin knockdown might be related to the inhibition of MYC and KRAS signals but the activation of interferon signals, inflammatory signals and p53 pathways.


Subject(s)
Adenocarcinoma of Lung , Cell Movement , Cell Proliferation , Lung Neoplasms , Neoplasm Invasiveness , Humans , Cell Proliferation/physiology , Cell Movement/physiology , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Disease Progression , A549 Cells , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic
9.
PLoS One ; 19(7): e0305396, 2024.
Article in English | MEDLINE | ID: mdl-38980840

ABSTRACT

The ADAMTS (a disintegrin and metalloprotease with thrombospondin motifs) family metalloprotease MIG-17 plays a crucial role in the migration of gonadal distal tip cells (DTCs) in Caenorhabditis elegans. MIG-17 is secreted from the body wall muscle cells and localizes to the basement membranes (BMs) of various tissues including the gonadal BM where it regulates DTC migration through its catalytic activity. Missense mutations in the BM protein genes, let-2/collagen IV a2 and fbl-1/fibulin-1, have been identified as suppressors of the gonadal defects observed in mig-17 mutants. Genetic analyses indicate that LET-2 and FBL-1 act downstream of MIG-17 to regulate DTC migration. In addition to the control of DTC migration, MIG-17 also plays a role in healthspan, but not in lifespan. Here, we examined whether let-2 and fbl-1 alleles can suppress the age-related phenotypes of mig-17 mutants. let-2(k196) fully and fbl-1(k201) partly, but not let-2(k193) and fbl-1(k206), suppressed the senescence defects of mig-17. Interestingly, fbl-1(k206), but not fbl-1(k201) or let-2 alleles, exhibited an extended lifespan compared to the wild type when combined with mig-17. These results reveal allele specific interactions between let-2 or fbl-1 and mig-17 in age-related phenotypes, indicating that basement membrane physiology plays an important role in organismal aging.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Collagen Type IV , Mutation , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Collagen Type IV/metabolism , Collagen Type IV/genetics , Longevity/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Basement Membrane/metabolism , Phenotype , Cell Movement/genetics , Gonads/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Disintegrins
10.
Sci Adv ; 10(25): eadn2332, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38896625

ABSTRACT

Satisfactory healing following acute tendon injury is marred by fibrosis. Despite the high frequency of tendon injuries and poor outcomes, there are no pharmacological therapies in use to enhance the healing process. Moreover, systemic treatments demonstrate poor tendon homing, limiting the beneficial effects of potential tendon therapeutics. To address this unmet need, we leveraged our existing tendon healing spatial transcriptomics dataset and identified an area enriched for expression of Acp5 (TRAP) and subsequently demonstrated robust TRAP activity in the healing tendon. This unexpected finding allowed us to refine and apply our existing TRAP binding peptide (TBP) functionalized nanoparticle (NP) drug delivery system (DDS) to facilitate improved delivery of systemic treatments to the healing tendon. To demonstrate the translational potential of this DDS, we delivered niclosamide (NEN), an S100a4 inhibitor. While systemic delivery of free NEN did not alter healing, TBP-NPNEN enhanced both functional and mechanical recovery, demonstrating the translational potential of this approach to enhance the tendon healing process.


Subject(s)
Tendon Injuries , Tendons , Wound Healing , Animals , Wound Healing/drug effects , Tendon Injuries/drug therapy , Tendons/drug effects , Tendons/metabolism , Drug Delivery Systems , Nanoparticles/chemistry , Mice , Nanoparticle Drug Delivery System/chemistry , Disease Models, Animal , Calcium-Binding Proteins/metabolism , Humans
11.
Retrovirology ; 21(1): 11, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38945996

ABSTRACT

BACKGROUND: Since the introduction of combination antiretroviral therapy (cART) the brain has become an important human immunodeficiency virus (HIV) reservoir due to the relatively low penetration of many drugs utilized in cART into the central nervous system (CNS). Given the inherent limitations of directly assessing acute HIV infection in the brains of people living with HIV (PLWH), animal models, such as humanized mouse models, offer the most effective means of studying the effects of different viral strains and their impact on HIV infection in the CNS. To evaluate CNS pathology during HIV-1 infection in the humanized bone marrow/liver/thymus (BLT) mouse model, a histological analysis was conducted on five CNS regions, including the frontal cortex, hippocampus, striatum, cerebellum, and spinal cord, to delineate the neuronal (MAP2ab, NeuN) and neuroinflammatory (GFAP, Iba-1) changes induced by two viral strains after 2 weeks and 8 weeks post-infection. RESULTS: Findings reveal HIV-infected human cells in the brain of HIV-infected BLT mice, demonstrating HIV neuroinvasion. Further, both viral strains, HIV-1JR-CSF and HIV-1CH040, induced neuronal injury and astrogliosis across all CNS regions following HIV infection at both time points, as demonstrated by decreases in MAP2ab and increases in GFAP fluorescence signal, respectively. Importantly, infection with HIV-1JR-CSF had more prominent effects on neuronal health in specific CNS regions compared to HIV-1CH040 infection, with decreasing number of NeuN+ neurons, specifically in the frontal cortex. On the other hand, infection with HIV-1CH040 demonstrated more prominent effects on neuroinflammation, assessed by an increase in GFAP signal and/or an increase in number of Iba-1+ microglia, across CNS regions. CONCLUSION: These findings demonstrate that CNS pathology is widespread during acute HIV infection. However, neuronal loss and the magnitude of neuroinflammation in the CNS is strain dependent indicating that strains of HIV cause differential CNS pathologies.


Subject(s)
Disease Models, Animal , HIV Infections , HIV-1 , Neuroinflammatory Diseases , Neurons , Animals , Mice , HIV Infections/virology , HIV Infections/pathology , HIV Infections/complications , Humans , Neurons/virology , Neurons/pathology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , Brain/pathology , Brain/virology , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Microfilament Proteins/metabolism
12.
Bull Exp Biol Med ; 176(6): 736-742, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38907060

ABSTRACT

Intranasal administration of total bovine brain gangliosides (6 mg/kg) to rats protected the CA1 hippocampal neurons from the death caused by two-vessel occlusion model (with hypotension) of forebrain ischemia/reperfusion injury. The immunohistochemical reaction of specific antibodies to marker proteins of activated microglia (Iba1) and astrocytes (GFAP) in hippocampal slices revealed the neuroprotective effect of exogenous gangliosides which can be mostly explained by their ability to suppress neuroinflammation and gliosis. The expression of neurotrophic factor BDNF in the CA1 region of hippocampus did not differ in sham-operated rats and animals exposed to ischemia/reperfusion. However, the administration of gangliosides increased the BDNF expression in both control and ischemic groups. The intranasal route of administration allows using lower concentrations of gangliosides preventing the death of hippocampal neurons.


Subject(s)
Administration, Intranasal , Brain-Derived Neurotrophic Factor , CA1 Region, Hippocampal , Gangliosides , Neurons , Neuroprotective Agents , Reperfusion Injury , Animals , Reperfusion Injury/pathology , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Gangliosides/pharmacology , Rats , Male , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/administration & dosage , Rats, Wistar , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Microfilament Proteins/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Brain Ischemia/metabolism , Prosencephalon/drug effects , Prosencephalon/pathology , Prosencephalon/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Cell Survival/drug effects , Disease Models, Animal
13.
Front Biosci (Landmark Ed) ; 29(6): 214, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38940025

ABSTRACT

BACKGROUND: The senescence marker protein 30 (SMP30) is a calcium-binding protein whose expression decreases with age, and is closely associated with hepatocellular carcinoma (HCC) development. The primary goal of this study was to examine the mechanistic effect of SMP30 on HCC migration and invasion. METHODS: Bioinformatic and immunohistochemical approaches were used to examine the expression of SMP30 in HCC tissues and its relationship to patient survival. We investigated the effects of SMP30 expression on HCC cell proliferation, migration, invasion, and cell cycle dynamics. cDNA microarray technology was used to determine the gene expression profile of SK-Hep-1 cells following recombinant SMP30 overexpression to identify genes downstream of SMP30 that regulate HCC cell migration and invasion. We identified SMP30 interacting proteins by affinity purification-mass spectrometry (AP-MS) and co-immunoprecipitation/western blotting (COIP-WB). RESULTS: SMP30 expression was lower in HCC tissues compared with normal liver tissues, and its expression positively correlated with overall survival in HCC patients. Additionally, SMP30 overexpression effectively blocked the migratory and invasive properties of SK-Hep-1 cells, but did not affect either proliferation rates or cell cycle. cDNA microarray results confirmed that many of the differentially expressed genes identified are involved in the process of epithelial-mesenchymal transition (EMT). AP-MS and COIP-WB experiments confirmed that Rho-associated protein kinase 1 (ROCK1) interacts with SMP30 in SK-Hep-1 cells, and ROCK1 is known to intimately regulate the EMT process. CONCLUSION: SMP30 inhibits HCC metastasis by influencing the expression of EMT-related proteins after interacting with ROCK1.


Subject(s)
Calcium-Binding Proteins , Carcinoma, Hepatocellular , Cell Movement , Epithelial-Mesenchymal Transition , Liver Neoplasms , Neoplasm Invasiveness , rho-Associated Kinases , Humans , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Epithelial-Mesenchymal Transition/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Female , Gene Expression Regulation, Neoplastic
14.
Sci Rep ; 14(1): 14734, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38926520

ABSTRACT

Based on the auditory periphery and the small head size, Etruscan shrews (Suncus etruscus) approximate ancestral mammalian conditions. The auditory brainstem in this insectivore has not been investigated. Using labelling techniques, we assessed the structures of their superior olivary complex (SOC) and the nuclei of the lateral lemniscus (NLL). There, we identified the position of the major nuclei, their input pattern, transmitter content, expression of calcium binding proteins (CaBPs) and two voltage-gated ion channels. The most prominent SOC structures were the medial nucleus of the trapezoid body (MNTB), the lateral nucleus of the trapezoid body (LNTB), the lateral superior olive (LSO) and the superior paraolivary nucleus (SPN). In the NLL, the ventral (VNLL), a specific ventrolateral VNLL (VNLLvl) cell population, the intermediate (INLL) and dorsal (DNLL) nucleus, as well as the inferior colliculus's central aspect were discerned. INLL and VNLL were clearly separated by the differential distribution of various marker proteins. Most labelled proteins showed expression patterns comparable to rodents. However, SPN neurons were glycinergic and not GABAergic and the overall CaBPs expression was low. Next to the characterisation of the Etruscan shrew's auditory brainstem, our work identifies conserved nuclei and indicates variable structures in a species that approximates ancestral conditions.


Subject(s)
Shrews , Superior Olivary Complex , Animals , Shrews/anatomy & histology , Superior Olivary Complex/anatomy & histology , Superior Olivary Complex/metabolism , Auditory Pathways/anatomy & histology , Neurons/metabolism , Inferior Colliculi/anatomy & histology , Inferior Colliculi/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Brain Stem/anatomy & histology , Brain Stem/metabolism , Male , Olivary Nucleus/anatomy & histology , Olivary Nucleus/metabolism
15.
Cells ; 13(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38920632

ABSTRACT

Non-coding RNAs (ncRNAs) have emerged as pivotal regulators in cellular biology, dispelling their former perception as 'junk transcripts'. Notably, the DLK1-DIO3 region harbors numerous ncRNAs, including long non-coding RNAs (lncRNAs) and over 50 microRNA genes. While papillary thyroid cancer showcases a pervasive decrease in DLK1-DIO3-derived ncRNA expression, the precise mechanisms driving this alteration remain elusive. We hypothesized that epigenetic alterations underlie shifts in ncRNA expression during thyroid cancer initiation and progression. This study aimed to elucidate the epigenetic mechanisms governing DLK1-DIO3 region expression in this malignancy. We have combined the analysis of DNA methylation by bisulfite sequencing together with that of histone modifications through ChIP-qPCR to gain insights into the epigenetic contribution to thyroid cancer in cell lines representing malignancies with different genetic backgrounds. Our findings characterize the region's epigenetic signature in thyroid cancer, uncovering distinctive DNA methylation patterns, particularly within CpG islands on the lncRNA MEG3-DMR, which potentially account for its downregulation in tumors. Pharmacological intervention targeting DNA methylation combined with histone deacetylation restored ncRNA expression. These results contribute to the understanding of the epigenetic mechanisms controlling the DLK1-DIO3 region in thyroid cancer, highlighting the combined role of DNA methylation and histone marks in regulating the locus' expression.


Subject(s)
Calcium-Binding Proteins , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Iodide Peroxidase , RNA, Long Noncoding , Thyroid Neoplasms , Humans , DNA Methylation/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , Cell Line, Tumor , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , CpG Islands/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Histones/metabolism , Membrane Proteins
16.
Cell Calcium ; 121: 102910, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823350

ABSTRACT

In cardiac myocytes, the type 2a sarco/endoplasmic reticulum Ca-ATPase (SERCA2a) plays a key role in intracellular Ca regulation. Due to its critical role in heart function, SERCA2a activity is tightly regulated by different mechanisms, including micropeptides. While phospholamban (PLB) is a well-known SERCA2a inhibitor, dwarf open reading frame (DWORF) is a recently identified SERCA2a activator. Since PLB phosphorylation is the most recognized mechanism of SERCA2a activation during adrenergic stress, we studied whether PLB phosphorylation also affects SERCA2a regulation by DWORF. By using confocal Ca imaging in a HEK293 expressing cell system, we analyzed the effect of the co-expression of PLB and DWORF using a bicistronic construct on SERCA2a-mediated Ca uptake. Under these conditions of matched expression of PLB and DWORF, we found that SERCA2a inhibition by non-phosphorylated PLB prevails over DWORF activating effect. However, when PLB is phosphorylated at PKA and CaMKII sites, not only PLB's inhibitory effect was relieved, but SERCA2a was effectively activated by DWORF. Förster resonance energy transfer (FRET) analysis between SERCA2a and DWORF showed that DWORF has a higher relative affinity for SERCA2a when PLB is phosphorylated. Thus, SERCA2a regulation by DWORF responds to the PLB phosphorylation status, suggesting that DWORF might contribute to SERCA2a activation during conditions of adrenergic stress.


Subject(s)
Calcium-Binding Proteins , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Calcium-Binding Proteins/metabolism , Humans , Phosphorylation , HEK293 Cells , Open Reading Frames/genetics , Calcium/metabolism , Enzyme Activation , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism
17.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38876797

ABSTRACT

Calcium is critical for regulating the waveform of motile cilia and flagella. Calaxin is currently the only known molecule involved in the calcium-dependent regulation in ascidians. We have recently shown that Calaxin stabilizes outer arm dynein (OAD), and the knockout of Calaxin results in primary ciliary dyskinesia phenotypes in vertebrates. However, from the knockout experiments, it was not clear which functions depend on calcium and how Calaxin regulates the waveform. To address this question, here, we generated transgenic zebrafish expressing a mutant E130A-Calaxin deficient in calcium binding. E130A-Calaxin restored the OAD reduction of calaxin -/- sperm and the abnormal movement of calaxin -/- left-right organizer cilia, showing that Calaxin's stabilization of OADs is calcium-independent. In contrast, our quantitative analysis of E130A-Calaxin sperms showed that the calcium-induced asymmetric beating was not restored, linking Calaxin's calcium-binding ability with an asymmetric flagellar beating for the first time. Our data show that Calaxin is a calcium-dependent regulator of the ciliary beating and a calcium-independent OAD stabilizer.


Subject(s)
Calcium-Binding Proteins , Spermatozoa , Zebrafish Proteins , Zebrafish , Animals , Male , Animals, Genetically Modified , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cilia/metabolism , Dyneins/metabolism , Dyneins/genetics , Flagella/metabolism , Flagella/physiology , Sperm Motility/genetics , Sperm Motility/physiology , Spermatozoa/metabolism , Spermatozoa/physiology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Cytoskeletal Proteins/metabolism
18.
Biomolecules ; 14(6)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38927078

ABSTRACT

Risk of cardiovascular disease mortality rises in women after menopause. While increased cardiovascular risk is largely attributed to postmenopausal declines in estrogens, the molecular changes in the heart that contribute to risk are poorly understood. Disruptions in intracellular calcium handling develop in ovariectomized mice and have been implicated in cardiac dysfunction. Using a mouse model of menopause in which ovarian failure occurs over 120 days, we sought to determine if perimenopause impacted calcium removal mechanisms in the heart and identify the molecular mechanisms. Mice were injected with 4-vinylcyclohexene diepoxide (VCD) to induce ovarian failure over 120 days, mimicking perimenopause. Hearts were removed at 60 and 120 days after VCD injections, representing the middle and end of perimenopause. SERCA2a function was significantly diminished at the end of perimenopause. Neither SERCA2a nor phospholamban expression changed at either time point, but phospholamban phosphorylation at S16 and T17 was dynamically altered. Intrinsic SERCA inhibitors sarcolipin and myoregulin increased >4-fold at day 60, as did the native activator DWORF. At the end of perimenopause, sarcolipin and myoregulin returned to baseline levels while DWORF was significantly reduced below controls. Sodium-calcium exchanger expression was significantly increased at the end of perimenopause. These results show that the foundation for increased cardiovascular disease mortality develops in the heart during perimenopause and that regulators of calcium handling exhibit significant fluctuations over time. Understanding the temporal development of cardiovascular risk associated with menopause and the underlying mechanisms is critical to developing interventions that mitigate the rise in cardiovascular mortality that arises after menopause.


Subject(s)
Disease Models, Animal , Perimenopause , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Female , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Mice , Perimenopause/metabolism , Vinyl Compounds/pharmacology , Myocardium/metabolism , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Primary Ovarian Insufficiency/metabolism , Cyclohexenes/pharmacology , Mice, Inbred C57BL , Phosphorylation
19.
Int Immunopharmacol ; 137: 112522, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38908089

ABSTRACT

BACKGROUND: Periodontitis is a common oral disease with high prevalence worldwide. Neural epidermal growth factor-like 1 protein (Nell-1) has recently been reported to have anti-inflammation effects and may be a drug candidate for osteoarthritis. However, its immunotherapeutic effects in periodontitis remain unknown. Therefore, this study aimed to investigate the effects of Nell-1 on periodontitis in terms of macrophage polarization and analyze its possible underlying mechanism. METHODS: A rat ligation-induced experimental periodontitis model was established and locally injected with Nell-1 (n = 6/group). Periodontal tissue destruction and macrophage polarization in vivo were analyzed using micro-CT, histology analysis, and western blot. Enzyme-linked immunosorbent assay was used to evaluate serum inflammatory cytokines. Then, the RAW 264.7 macrophage cells were treated with lipopolysaccharide (LPS), Nell-1, and the c-Jun N-terminal kinases (JNK) inhibitor (SP600125). RT-PCR, western blot, and flow cytometry were performed to further analyze the effect of Nell-1 on macrophage polarization and the underlying mechanism in vitro. RESULTS: Local treatment with Nell-1 significantly alleviated the destruction of alveolar bone and fibers in periodontitis, and upregulated the ratio of M2/M1 macrophages in periodontal tissues (P < 0.05). In vitro, Nell-1 at the concentrations of 200 and 500 ng/mL could significantly inhibit the expression of M1-related inflammatory factors in LPS-stimulated macrophages, and increase the expression of M2-related markers, regulating the macrophage phenotype switch into M2 (P < 0.05). The mRNA of JNK and relative protein level of phospho-JNK/JNK were also upregulated by Nell-1 (P < 0.05). Additionally, the JNK inhibitor (SP600125) could reverse the effect of Nell-1 on macrophage polarization (P < 0.05). CONCLUSIONS: Nell-1 could modulate the ratio of M2/M1 macrophages possibly through the JNK/MAPK signaling pathway, subsequently attenuating the inflammation and destruction of periodontal tissues caused by periodontitis.


Subject(s)
Macrophages , Periodontitis , Animals , Periodontitis/drug therapy , Periodontitis/immunology , Periodontitis/pathology , Periodontitis/metabolism , Mice , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , RAW 264.7 Cells , Rats , Male , Cytokines/metabolism , Rats, Sprague-Dawley , Lipopolysaccharides , Disease Models, Animal , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophage Activation/drug effects , Phenotype , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics
20.
Nat Commun ; 15(1): 5188, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898014

ABSTRACT

Autophagy is relevant for diverse processes in eukaryotic cells, making its regulation of fundamental importance. The formation and maturation of autophagosomes require a complex choreography of numerous factors. The endosomal sorting complex required for transport (ESCRT) is implicated in the final step of autophagosomal maturation by sealing of the phagophore membrane. ESCRT-III components were shown to mediate membrane scission by forming filaments that interact with cellular membranes. However, the molecular mechanisms underlying the recruitment of ESCRTs to non-endosomal membranes remain largely unknown. Here we focus on the ESCRT-associated protein ALG2-interacting protein X (ALIX) and identify Ca2+-dependent lipid binding protein 1 (CaLB1) as its interactor. Our findings demonstrate that CaLB1 interacts with AUTOPHAGY8 (ATG8) and PI(3)P, a phospholipid found in autophagosomal membranes. Moreover, CaLB1 and ALIX localize with ATG8 on autophagosomes upon salt treatment and assemble together into condensates. The depletion of CaLB1 impacts the maturation of salt-induced autophagosomes and leads to reduced delivery of autophagosomes to the vacuole. Here, we propose a crucial role of CaLB1 in augmenting phase separation of ALIX, facilitating the recruitment of ESCRT-III to the site of phagophore closure thereby ensuring efficient maturation of autophagosomes.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Autophagosomes , Autophagy , Calcium-Binding Proteins , Endosomal Sorting Complexes Required for Transport , Arabidopsis/metabolism , Arabidopsis/genetics , Autophagosomes/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Phosphatidylinositol Phosphates/metabolism , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Protein 8 Family/genetics , Vacuoles/metabolism , Phase Separation
SELECTION OF CITATIONS
SEARCH DETAIL
...