Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.275
Filter
1.
Gen Physiol Biophys ; 43(4): 321-333, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953574

ABSTRACT

Vitiligo is featured by manifestation of white maculae and primarily results from oxidative stress. Sphingosine kinase-1 (SPHK1) participates in oxidative stress. This paper was devised to explore the role of SPHK1 in vitiligo and to disclose the mechanism. PIG1 cell viability was appraised utilizing cell counting kit-8 assay while Western blot detected SPHK1 and four and a half LIM domains 2 (FHL2). The transduction efficacy of small interfering RNA (siRNA)-SPHK1, siRNA-FHL2 and pcDNA3.1 plasmid overexpressing FHL2 (Ov-FHL2) was checked using Western blot. Flow cytometry detected cell apoptotisis. Western blot detected mitochondrial cytochrome c (Mit-Cyt-c) and cytosolic cytochrome c (Cyto-Cyt-c). Dichloro-dihydro-fluorescein diacetate (DCFH-DA) detected reactive oxygen species (ROS) activity while oxidative stress markers were evaluated using corresponding assay kits. SPHK1 expression was discovered to be increased in hydrogen peroxide (H2O2)-challenged PIG1 cells and SPHK1 interference alleviated H2O2-challenged viability damage, apoptosis, oxidative stress and FHL2 expression in PIG1 cells. FHL2 depletion could suppress viability damage, apoptosis and oxidative stress in H2O2-challenged PIG1 cells. Rescue experiments demonstrated that the suppressive impacts of SPHK1 deficiency on PIG1 cell viability, apoptosis and oxidative stress induced by H2O2 were offset by FHL2 overexpression. Collectively, SPHK1 knockdown protected against vitiligo via the regulation of FHL2.


Subject(s)
Cell Survival , Hydrogen Peroxide , LIM-Homeodomain Proteins , Melanocytes , Oxidative Stress , Phosphotransferases (Alcohol Group Acceptor) , Oxidative Stress/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Hydrogen Peroxide/metabolism , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Humans , Melanocytes/metabolism , Melanocytes/drug effects , Cell Survival/drug effects , Apoptosis/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Cell Line
2.
Cell Mol Life Sci ; 81(1): 286, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38970652

ABSTRACT

Paralog factors are considered to ensure the robustness of biological processes by providing redundant activity in cells where they are co-expressed. However, the specific contribution of each factor is frequently underestimated. In the developing spinal cord, multiple families of transcription factors successively contribute to differentiate an initially homogenous population of neural progenitors into a myriad of neuronal subsets with distinct molecular, morphological, and functional characteristics. The LIM-homeodomain transcription factors Lhx3, Lhx4, Isl1 and Isl2 promote the segregation and differentiation of spinal motor neurons and V2 interneurons. Based on their high sequence identity and their similar distribution, the Lhx3 and Lhx4 paralogs are considered to contribute similarly to these processes. However, the specific contribution of Lhx4 has never been studied. Here, we provide evidence that Lhx3 and Lhx4 are present in the same cell populations during spinal cord development. Similarly to Lhx3, Lhx4 can form multiproteic complexes with Isl1 or Isl2 and the nuclear LIM interactor NLI. Lhx4 can stimulate a V2-specific enhancer more efficiently than Lhx3 and surpasses Lhx3 in promoting the differentiation of V2a interneurons in chicken embryo electroporation experiments. Finally, Lhx4 inactivation in mice results in alterations of differentiation of the V2a subpopulation, but not of motor neuron production, suggesting that Lhx4 plays unique roles in V2a differentiation that are not compensated by the presence of Lhx3. Thus, Lhx4 could be the major LIM-HD factor involved in V2a interneuron differentiation during spinal cord development and should be considered for in vitro differentiation of spinal neuronal populations.


Subject(s)
Cell Differentiation , Interneurons , LIM-Homeodomain Proteins , Spinal Cord , Transcription Factors , Animals , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Interneurons/metabolism , Interneurons/cytology , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord/embryology , Chick Embryo , Mice , Motor Neurons/metabolism , Motor Neurons/cytology , Humans , Gene Expression Regulation, Developmental
3.
Genes (Basel) ; 15(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38927675

ABSTRACT

Lhx3 is a LIM-homeodomain transcription factor that affects body size in mammals by regulating the secretion of pituitary hormones. Akita, Shiba Inu, and Mame Shiba Inu dogs are Japanese native dog breeds that have different body sizes. To determine whether Lhx3 plays a role in the differing body sizes of these three dog breeds, we sequenced the Lhx3 gene in the three breeds, which led to the identification of an SNP in codon 280 (S280N) associated with body size. The allele frequency at this SNP differed significantly between the large Akita and the two kinds of smaller Shiba dogs. To validate the function of this SNP on body size, we introduced this change into the Lhx3 gene of mice. Homozygous mutant mice (S279N+/+) were found to have significantly increased body lengths and weights compared to heterozygous mutant (S279N+/-) and wild-type (S279N-/-) mice several weeks after weaning. These results demonstrate that a nonsynonymous substitution in Lhx3 plays an important role in regulating body size in mammals.


Subject(s)
Body Size , LIM-Homeodomain Proteins , Polymorphism, Single Nucleotide , Transcription Factors , Animals , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Transcription Factors/genetics , Mice , Body Size/genetics , Dogs/genetics , Gene Frequency , Male , Female
4.
Cardiovasc Toxicol ; 24(7): 646-655, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38801481

ABSTRACT

This research is concentrated on investigating the role and mechanism of miR-652-3p in the protective effects of isoflurane (ISO) against myocardial ischemia-reperfusion (I/R) injury. H9c2 cells underwent pretreatment with varying concentrations of ISO, and subsequently, a hypoxia/reoxygenation (H/R) model was constructed. The levels of miR-652-3p, ISL LIM homeobox 1 (ISL1), and inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-alpha (TNF-α) were evaluated through reverse transcription polymerase chain reaction (RT-qPCR). Enzyme-linked immunosorbent assay was employed to investigate concentrations of myocardial injury markers, such as creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI). Cell counting kit-8 was used to evaluate cell viability, while flow cytometry was utilized to measure apoptosis. Additionally, a dual luciferase reporter assay was conducted to validate the targeting relationship between ISL1 and miR-652-3p. Herein, we confirmed that the level of miR-652-3p was gradually increased with prolonged hypoxia; nevertheless, this increase was suppressed by ISO pretreatment (P < 0.05). Additionally, ISO pretreatment prevented the decrease in cell viability, increase in apoptosis, and overproduction of IL-6, TNF-α, CK-MB, and cTnI induced by H/R (P < 0.05). However, the inhibitory effects of ISO were counteracted by the increased levels of miR-652-3p (P < 0.05). ISL1 is a potential target of miR-652-3p. H/R induction suppressed ISL1 levels compared to the control, but ISO treatment increased its expression (P < 0.05). Overexpression of ISL1 inhibited the elimination of the protective effect of ISO on myocardial damage induced by the elevation of miR-652-3p (P < 0.05). The findings of this research confirm that miR-652-3p attenuated the protective effect of ISO on cardiomyocytes in myocardial ischemia by targeting ISL1.


Subject(s)
Apoptosis , Cell Hypoxia , Interleukin-6 , Isoflurane , LIM-Homeodomain Proteins , MicroRNAs , Myocardial Reperfusion Injury , Myocytes, Cardiac , Transcription Factors , MicroRNAs/metabolism , MicroRNAs/genetics , Isoflurane/pharmacology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/genetics , Cell Line , Apoptosis/drug effects , Rats , Transcription Factors/metabolism , Transcription Factors/genetics , Interleukin-6/metabolism , Interleukin-6/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Creatine Kinase, MB Form/metabolism , Creatine Kinase, MB Form/blood , Troponin I/metabolism , Cytoprotection
5.
Tissue Cell ; 88: 102387, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703583

ABSTRACT

Stem cell-mediated tissue regeneration is a promising strategy for repairing tissue defects and functional reconstruction in periodontitis, a common disease that leads to the loss of alveolar bone and teeth. However, stem cell apoptosis, widely observed during tissue regeneration, impairs its efficiency. Therefore, the regulation of stem cell apoptosis is critical for improving regeneration efficiency. The LIM homeobox 8 gene LHX8, belongs to the LIM homeobox family, which was involved in tooth morphogenesis. Here, we found that LHX8 was significantly expressed in dental pulp. LHX8 knockdown significantly increased dental pulp mesenchymal stem cells (DPSCs) apoptosis, as confirmed by RT-PCR, western blotting, flow cytometry, and transmission electron microscopy. Additionally, LHX8 overexpression inhibited apoptosis and enhanced the osteo/odontogenic differentiation potential of hDPSCs in vitro. Furthermore, LHX8-overexpression could enhance the periodontal tissue regeneration efficiency of hDPSCs in mice with periodontitis. In conclusion, the present study indicates that LHX8 inhibits stem cell apoptosis and promotes functional tissue formation in stem cell-based tissue regeneration engineering, suggesting a new therapeutic target to increase the efficacy of periodontal tissue regeneration.


Subject(s)
Apoptosis , Dental Pulp , LIM-Homeodomain Proteins , Regeneration , Transcription Factors , Dental Pulp/cytology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , Mice , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Differentiation/genetics , Stem Cells/metabolism , Stem Cells/cytology , Periodontium
6.
Cell Rep Med ; 5(5): 101554, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38729157

ABSTRACT

The axons of retinal ganglion cells (RGCs) form the optic nerve, transmitting visual information from the eye to the brain. Damage or loss of RGCs and their axons is the leading cause of visual functional defects in traumatic injury and degenerative diseases such as glaucoma. However, there are no effective clinical treatments for nerve damage in these neurodegenerative diseases. Here, we report that LIM homeodomain transcription factor Lhx2 promotes RGC survival and axon regeneration in multiple animal models mimicking glaucoma disease. Furthermore, following N-methyl-D-aspartate (NMDA)-induced excitotoxicity damage of RGCs, Lhx2 mitigates the loss of visual signal transduction. Mechanistic analysis revealed that overexpression of Lhx2 supports axon regeneration by systematically regulating the transcription of regeneration-related genes and inhibiting transcription of Semaphorin 3C (Sema3C). Collectively, our studies identify a critical role of Lhx2 in promoting RGC survival and axon regeneration, providing a promising neural repair strategy for glaucomatous neurodegeneration.


Subject(s)
Axons , Disease Models, Animal , Glaucoma , LIM-Homeodomain Proteins , Nerve Regeneration , Retinal Ganglion Cells , Transcription Factors , Animals , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Glaucoma/genetics , Glaucoma/pathology , Glaucoma/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Axons/metabolism , Axons/pathology , Mice , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Mice, Inbred C57BL , Cell Survival/genetics , Semaphorins/metabolism , Semaphorins/genetics , N-Methylaspartate/metabolism
7.
Nat Commun ; 15(1): 4496, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802383

ABSTRACT

Titin N2B unique sequence (N2B-us) is a 572 amino acid sequence that acts as an elastic spring to regulate muscle passive elasticity. It is thought to lack stable tertiary structures and is a force-bearing region that is regulated by mechanical stretching. In this study, the conformation of N2B-us and its interaction with four-and-a-half LIM domain protein 2 (FHL2) are investigated using AlphaFold2 predictions and single-molecule experimental validation. Surprisingly, a stable alpha/beta structural domain is predicted and confirmed in N2B-us that can be mechanically unfolded at forces of a few piconewtons. Additionally, more than twenty FHL2 LIM domain binding sites are predicted to spread throughout N2B-us. Single-molecule manipulation experiments reveals the force-dependent binding of FHL2 to the N2B-us structural domain. These findings provide insights into the mechano-sensing functions of N2B-us and its interactions with FHL2.


Subject(s)
Connectin , LIM-Homeodomain Proteins , Protein Binding , Protein Domains , Transcription Factors , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/chemistry , LIM-Homeodomain Proteins/genetics , Connectin/metabolism , Connectin/chemistry , Connectin/genetics , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Binding Sites , Humans , Animals , Muscle Proteins/metabolism , Muscle Proteins/chemistry , Muscle Proteins/genetics , Amino Acid Sequence
8.
Stem Cell Reports ; 19(6): 830-838, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38759646

ABSTRACT

The differentiation of human pluripotent stem cells into ventral mesencephalic dopaminergic (DA) fate is relevant for the treatment of Parkinson's disease. Shortcuts to obtaining DA cells through direct reprogramming often include forced expression of the transcription factor LMX1A. Although reprogramming with LMX1A can generate tyrosine hydroxylase (TH)-positive cells, their regional identity remains elusive. Using an in vitro model of early human neural tube patterning, we report that forced LMX1A expression induced a ventral-to-dorsal fate shift along the entire neuroaxis with the emergence of roof plate fates despite the presence of ventralizing molecules. The LMX1A-expressing progenitors gave rise to grafts containing roof plate-derived choroid plexus cysts as well as ectopically induced TH-positive neurons of a forebrain identity. Early activation of LMX1A prior to floor plate specification was necessary for the dorsalizing effect. Our work suggests using caution in employing LMX1A for the induction of DA fate, as this factor may generate roof plate rather than midbrain fates.


Subject(s)
Cell Differentiation , Dopaminergic Neurons , Human Embryonic Stem Cells , LIM-Homeodomain Proteins , Mesencephalon , Transcription Factors , Humans , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/cytology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Mesencephalon/cytology , Mesencephalon/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Body Patterning/genetics , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/genetics , Animals , Gene Expression Regulation, Developmental
9.
Cell Mol Life Sci ; 81(1): 244, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38814462

ABSTRACT

Four-and-a-half LIM domains protein 2 (FHL2) is an adaptor protein that may interact with hypoxia inducible factor 1α (HIF-1α) or ß-catenin, two pivotal protective signaling in acute kidney injury (AKI). However, little is known about the regulation and function of FHL2 during AKI. We found that FHL2 was induced in renal tubular cells in patients with acute tubular necrosis and mice model of ischemia-reperfusion injury (IRI). In cultured renal proximal tubular cells (PTCs), hypoxia induced FHL2 expression and promoted the binding of HIF-1 to FHL2 promoter. Compared with control littermates, mice with PTC-specific deletion of FHL2 gene displayed worse renal function, more severe morphologic lesion, more tubular cell death and less cell proliferation, accompanying by downregulation of AQP1 and Na, K-ATPase after IRI. Consistently, loss of FHL2 in PTCs restricted activation of HIF-1 and ß-catenin signaling simultaneously, leading to attenuation of glycolysis, upregulation of apoptosis-related proteins and downregulation of proliferation-related proteins during IRI. In vitro, knockdown of FHL2 suppressed hypoxia-induced activation of HIF-1α and ß-catenin signaling pathways. Overexpression of FHL2 induced physical interactions between FHL2 and HIF-1α, ß-catenin, GSK-3ß or p300, and the combination of these interactions favored the stabilization and nuclear translocation of HIF-1α and ß-catenin, enhancing their mediated gene transcription. Collectively, these findings identify FHL2 as a direct downstream target gene of HIF-1 signaling and demonstrate that FHL2 could play a critical role in protecting against ischemic AKI by promoting the activation of HIF-1 and ß-catenin signaling through the interactions with its multiple protein partners.


Subject(s)
Acute Kidney Injury , Kidney Tubules, Proximal , LIM-Homeodomain Proteins , Muscle Proteins , Reperfusion Injury , Transcription Factors , beta Catenin , Animals , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/genetics , Mice , beta Catenin/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Male , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Signal Transduction , Mice, Inbred C57BL , Mice, Knockout , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Cell Proliferation , Apoptosis
10.
Sci Adv ; 10(21): eadk2149, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781326

ABSTRACT

Understanding the genetic programs that drive neuronal diversification into classes and subclasses is key to understand nervous system development. All neurons can be classified into two types: commissural and ipsilateral, based on whether their axons cross the midline or not. However, the gene regulatory program underlying this binary division is poorly understood. We identified a pair of basic helix-loop-helix transcription factors, Nhlh1 and Nhlh2, as a global transcriptional mechanism that controls the laterality of all floor plate-crossing commissural axons in mice. Mechanistically, Nhlh1/2 play an essential role in the expression of Robo3, the key guidance molecule for commissural axon projections. This genetic program appears to be evolutionarily conserved in chick. We further discovered that Isl1, primarily expressed in ipsilateral neurons within neural tubes, negatively regulates the Robo3 induction by Nhlh1/2. Our findings elucidate a gene regulatory strategy where a conserved global mechanism intersects with neuron class-specific regulators to control the partitioning of neurons based on axon laterality.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Gene Expression Regulation, Developmental , Neurons , Animals , Neurons/metabolism , Neurons/cytology , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Axons/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Chick Embryo , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Gene Regulatory Networks
11.
J Biol Chem ; 300(5): 107254, 2024 May.
Article in English | MEDLINE | ID: mdl-38569934

ABSTRACT

Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.


Subject(s)
Connectin , LIM Domain Proteins , Muscle Proteins , Myocytes, Cardiac , Nerve Tissue Proteins , Nuclear Proteins , Sarcomeres , Animals , Humans , Mice , Rats , Connectin/metabolism , Connectin/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Protein Binding , Sarcomeres/metabolism , Transcription Factors
12.
Cell Rep ; 43(5): 114157, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38678557

ABSTRACT

The sensory cortex receives synaptic inputs from both first-order and higher-order thalamic nuclei. First-order inputs relay simple stimulus properties from the periphery, whereas higher-order inputs relay more complex response properties, provide contextual feedback, and modulate plasticity. Here, we reveal that a cortical neuron's higher-order input is determined by the type of progenitor from which it is derived during embryonic development. Within layer 4 (L4) of the mouse primary somatosensory cortex, neurons derived from intermediate progenitors receive stronger higher-order thalamic input and exhibit greater higher-order sensory responses. These effects result from differences in dendritic morphology and levels of the transcription factor Lhx2, which are specified by the L4 neuron's progenitor type. When this mechanism is disrupted, cortical circuits exhibit altered higher-order responses and sensory-evoked plasticity. Therefore, by following distinct trajectories, progenitor types generate diversity in thalamocortical circuitry and may provide a general mechanism for differentially routing information through the cortex.


Subject(s)
Somatosensory Cortex , Thalamus , Transcription Factors , Animals , Mice , Thalamus/cytology , Thalamus/embryology , Thalamus/physiology , Transcription Factors/metabolism , Somatosensory Cortex/cytology , Somatosensory Cortex/physiology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Neurons/cytology , Neurons/physiology , Neurons/metabolism , Neuronal Plasticity/physiology , Mice, Inbred C57BL
13.
Mol Carcinog ; 63(7): 1334-1348, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38629424

ABSTRACT

Gastrointestinal stromal tumors (GISTs) are predominately induced by KIT mutants. In this study, we found that four and a half LIM domains 2 (FHL2) was highly expressed in GISTs and KIT signaling dramatically increased FHL2 transcription while FHL2 inhibited KIT transcription. In addition, our results showed that FHL2 associated with KIT and increased the ubiquitination of both wild-type KIT and primary KIT mutants in GISTs, leading to decreased expression and activation of KIT although primary KIT mutants were less inhibited by FHL2 than wild-type KIT. In the animal experiments, loss of FHL2 expression in mice carrying germline KIT/V558A mutation which can develop GISTs resulted in increased tumor growth, but increased sensitivity of GISTs to imatinib treatment which is used as the first-line targeted therapy of GISTs, suggesting that FHL2 plays a role in the response of GISTs to KIT inhibitor. Unlike wild-type KIT and primary KIT mutants, we further found that FHL2 didn't alter the expression and activation of drug-resistant secondary KIT mutants. Taken together, our results indicated that FHL2 acts as the negative feedback of KIT signaling in GISTs while primary KIT mutants are less sensitive and secondary KIT mutants are resistant to the inhibition of FHL2.


Subject(s)
Gastrointestinal Stromal Tumors , LIM-Homeodomain Proteins , Muscle Proteins , Proto-Oncogene Proteins c-kit , Signal Transduction , Transcription Factors , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gastrointestinal Stromal Tumors/metabolism , Animals , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Humans , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Mutation , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Imatinib Mesylate/pharmacology , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/metabolism , Cell Line, Tumor , Ubiquitination
14.
J Mol Cell Cardiol ; 191: 40-49, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38604403

ABSTRACT

The heart has the ability to detect and respond to changes in mechanical load through a process called mechanotransduction. In this study, we focused on investigating the role of the cardiac-specific N2B element within the spring region of titin, which has been proposed to function as a mechanosensor. To assess its significance, we conducted experiments using N2B knockout (KO) mice and wildtype (WT) mice, subjecting them to three different conditions: 1) cardiac pressure overload induced by transverse aortic constriction (TAC), 2) volume overload caused by aortocaval fistula (ACF), and 3) exercise-induced hypertrophy through swimming. Under conditions of pressure overload (TAC), both genotypes exhibited similar hypertrophic responses. In contrast, WT mice displayed robust left ventricular hypertrophy after one week of volume overload (ACF), while the KO mice failed to undergo hypertrophy and experienced a high mortality rate. Similarly, swim exercise-induced hypertrophy was significantly reduced in the KO mice. RNA-Seq analysis revealed an abnormal ß-adrenergic response to volume overload in the KO mice, as well as a diminished response to isoproterenol-induced hypertrophy. Because it is known that the N2B element interacts with the four-and-a-half LIM domains 1 and 2 (FHL1 and FHL2) proteins, both of which have been associated with mechanotransduction, we evaluated these proteins. Interestingly, while volume-overload resulted in FHL1 protein expression levels that were comparable between KO and WT mice, FHL2 protein levels were reduced by over 90% in the KO mice compared to WT. This suggests that in response to volume overload, FHL2 might act as a signaling mediator between the N2B element and downstream signaling pathways. Overall, our study highlights the importance of the N2B element in mechanosensing during volume overload, both in physiological and pathological settings.


Subject(s)
Connectin , Mechanotransduction, Cellular , Mice, Knockout , Animals , Mice , Connectin/metabolism , Connectin/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/genetics , Myocardium/metabolism , Myocardium/pathology , Male , Physical Conditioning, Animal , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Disease Models, Animal , Muscle Proteins/metabolism , Muscle Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Protein Kinases , Intracellular Signaling Peptides and Proteins
15.
Endocr Regul ; 58(1): 47-56, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38563293

ABSTRACT

OBJECTIVE.: Homeobox genes play an important role in health and disease including oncogenesis. The present investigation aimed to study ERN1-dependent hypoxic regulation of the expression of genes encoding homeobox proteins MEIS (zinc finger E-box binding homeobox 2) and LIM homeobox 1 family, SPAG4 (sperm associated antigen 4) and NKX3-1 (NK3 homeobox 1) in U87MG glioblastoma cells in response to inhibition of ERN1 (endoplasmic reticulum to nucleus signaling 1) for evaluation of their possible significance in the control of glioblastoma growth. METHODS.: The expression level of homeobox genes was studied in control (transfected by vector) and ERN1 knockdown U87MG glioblastoma cells under hypoxia induced by dimethyloxalylglycine (0.5 mM for 4 h) by quantitative polymerase chain reaction and normalized to ACTB. RESULTS.: It was found that hypoxia down-regulated the expression level of LHX2, LHX6, MEIS2, and NKX3-1 genes but up-regulated the expression level of MEIS1, LHX1, MEIS3, and SPAG4 genes in control glioblastoma cells. At the same time, ERN1 knockdown of glioblastoma cells significantly modified the sensitivity of all studied genes to a hypoxic condition. Thus, ERN1 knockdown of glioblastoma cells removed the effect of hypoxia on the expression of MEIS1 and LHX1 genes, but increased the sensitivity of MEIS2, LHX2, and LHX6 genes to hypoxia. However, the expression of MEIS3, NKX3-1, and SPAG4 genes had decreased sensitivity to hypoxia in ERN1 knockdown glioblastoma cells. Moreover, more pronounced changes under the conditions of ERN1 inhibition were detected for the pro-oncogenic gene SPAG4. CONCLUSION.: The results of the present study demonstrate that hypoxia affected the expression of homeobox genes MEIS1, MEIS2, MEIS3, LHX1, LHX2, LHX6, SPAG4, and NKX3-1 in U87MG glioblastoma cells in gene-specific manner and that the sensitivity of all studied genes to hypoxia condition is mediated by ERN1, the major pathway of the endoplasmic reticulum stress signaling, and possibly contributed to the control of glioblastoma growth. A fundamentally new results of this work is the establishment of the fact regarding the dependence of hypoxic regulation of SPAG4 gene expression on ER stress, in particular ERN1, which is associated with suppression of cell proliferation and tumor growth.


Subject(s)
Glioblastoma , Humans , Glioblastoma/genetics , Genes, Homeobox , Protein Serine-Threonine Kinases/genetics , LIM-Homeodomain Proteins/genetics , Cell Hypoxia/genetics , Gene Expression Regulation, Neoplastic/genetics , Hypoxia/genetics , Transcription Factors/genetics , Gene Expression , Cell Line, Tumor , Gene Knockdown Techniques , Endoribonucleases/genetics
16.
Stem Cell Res ; 76: 103376, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38452706

ABSTRACT

The ISL LIM homeobox 1 (ISL1) gene belongs to the LIM/homeodomain transcription factor family and plays a pivotal role in conveying multipotent and proliferative properties of cardiac precursor cells. Mutations in ISL1 are linked to congenital heart disease. To further explore ISL1's role in the human heart, we have created a homozygous ISL1 knockout (ISL1-KO) human embryonic stem cell line using the CRISPR/Cas9 system. Notably, this ISL1-KO cell line retains normal morphology, pluripotency, and karyotype. This resource serves as a valuable tool for investigating ISL1's function in cardiomyocyte differentiation.


Subject(s)
CRISPR-Cas Systems , Human Embryonic Stem Cells , Humans , CRISPR-Cas Systems/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Line , Heart , Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/metabolism , LIM-Homeodomain Proteins/genetics
17.
J Neurosci ; 44(18)2024 May 01.
Article in English | MEDLINE | ID: mdl-38438260

ABSTRACT

Locomotion allows us to move and interact with our surroundings. Spinal networks that control locomotion produce rhythm and left-right and flexor-extensor coordination. Several glutamatergic populations, Shox2 non-V2a, Hb9-derived interneurons, and, recently, spinocerebellar neurons have been proposed to be involved in the mouse rhythm generating networks. These cells make up only a smaller fraction of the excitatory cells in the ventral spinal cord. Here, we set out to identify additional populations of excitatory spinal neurons that may be involved in rhythm generation or other functions in the locomotor network. We use RNA sequencing from glutamatergic, non-glutamatergic, and Shox2 cells in the neonatal mice from both sexes followed by differential gene expression analyses. These analyses identified transcription factors that are highly expressed by glutamatergic spinal neurons and differentially expressed between Shox2 neurons and glutamatergic neurons. From this latter category, we identified the Lhx9-derived neurons as having a restricted spinal expression pattern with no Shox2 neuron overlap. They are purely glutamatergic and ipsilaterally projecting. Ablation of the glutamatergic transmission or acute inactivation of the neuronal activity of Lhx9-derived neurons leads to a decrease in the frequency of locomotor-like activity without change in coordination pattern. Optogenetic activation of Lhx9-derived neurons promotes locomotor-like activity and modulates the frequency of the locomotor activity. Calcium activities of Lhx9-derived neurons show strong left-right out-of-phase rhythmicity during locomotor-like activity. Our study identifies a distinct population of spinal excitatory neurons that regulates the frequency of locomotor output with a suggested role in rhythm-generation in the mouse alongside other spinal populations.


Subject(s)
Interneurons , LIM-Homeodomain Proteins , Locomotion , Spinal Cord , Transcription Factors , Animals , Interneurons/physiology , Mice , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Locomotion/physiology , Spinal Cord/physiology , Spinal Cord/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Male , Female , Glutamic Acid/metabolism , Animals, Newborn , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
18.
Medicine (Baltimore) ; 103(10): e37442, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38457557

ABSTRACT

BACKGROUND: Genetic factors contribute to chronic kidney disease (CKD) and end-stage renal disease (ESRD). Advances in genetic testing have enabled the identification of hereditary kidney diseases, including those caused by LMX1B mutations. LMX1B mutations can lead to nail-patella syndrome (NPS) or nail-patella-like renal disease (NPLRD) with only renal manifestations. CASE PRESENTATION: The proband was a 13-year-old female who was diagnosed with nephrotic syndrome at the age of 6. Then she began intermittent hormone and drug therapy. When she was 13 years old, she was admitted to our hospital due to sudden chest tightness, which progressed to end-stage kidney disease (ESRD), requiring kidney replacement therapy. Whole-Exome Sequencing (WES) results suggest the presence of LMX1B gene mutation, c.737G > T, p.Arg246Leu. Tracing her family history, we found that her father, grandmother, uncle and 2 cousins all had hematuria, or proteinuria. In addition to the grandmother, a total of 9 members of the family performed WES. The members with kidney involved all carry the mutated gene. Healthy members did not have the mutated gene. It is characterized by co-segregation of genotype and phenotype. We followed the family for 9 year, the father developed ESRD at the age of 50 and started hemodialysis treatment. The rest patients had normal renal function. No extra-renal manifestations associated with NPS were found in any member of the family. CONCLUSIONS: This study has successfully identified missense mutation, c.737G > T (p.Arg246Leu) in the homeodomain, which appears to be responsible for isolated nephropathy in the studied family. The arginine to leucine change at codon 246 likely disrupts the DNA-binding homeodomain of LMX1B. Previous research has documented 2 types of mutations at codon R246, namely R246Q and R246P, which are known to cause NPLRD. The newly discovered mutation, R246L, is likely to be another novel mutation associated with NPLRD, thus expanding the range of mutations at the crucial renal-critical codon 246 that contribute to the development of NPLRD. Furthermore, our findings suggest that any missense mutation occurring at the 246th amino acid position within the homeodomain of the LMX1B gene has the potential to lead to NPLRD.


Subject(s)
Kidney Failure, Chronic , Nail-Patella Syndrome , Nephritis, Hereditary , Humans , Female , Adolescent , Transcription Factors/genetics , LIM-Homeodomain Proteins/genetics , Nephritis, Hereditary/genetics , Mutation , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/therapy , Codon , China , Homeodomain Proteins/genetics
19.
Nat Neurosci ; 27(5): 862-872, 2024 May.
Article in English | MEDLINE | ID: mdl-38528203

ABSTRACT

The mammalian telencephalon contains distinct GABAergic projection neuron and interneuron types, originating in the germinal zone of the embryonic basal ganglia. How genetic information in the germinal zone determines cell types is unclear. Here we use a combination of in vivo CRISPR perturbation, lineage tracing and ChIP-sequencing analyses and show that the transcription factor MEIS2 favors the development of projection neurons by binding enhancer regions in projection-neuron-specific genes during mouse embryonic development. MEIS2 requires the presence of the homeodomain transcription factor DLX5 to direct its functional activity toward the appropriate binding sites. In interneuron precursors, the transcription factor LHX6 represses the MEIS2-DLX5-dependent activation of projection-neuron-specific enhancers. Mutations of Meis2 result in decreased activation of regulatory enhancers, affecting GABAergic differentiation. We propose a differential binding model where the binding of transcription factors at cis-regulatory elements determines differential gene expression programs regulating cell fate specification in the mouse ganglionic eminence.


Subject(s)
Embryonic Development , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins , Transcription Factors , Animals , Mice , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Embryonic Development/physiology , Enhancer Elements, Genetic/genetics , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Cell Differentiation/physiology , Interneurons/metabolism , Interneurons/physiology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Neurogenesis/physiology , Nerve Tissue Proteins
20.
FEBS J ; 291(13): 2792-2810, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38465368

ABSTRACT

LIM homeodomain transcription factor 1-alpha (LMX1a) is a neuronal lineage-specific transcription activator that plays an essential role during the development of midbrain dopaminergic (mDA) neurons. LMX1a induces the expression of multiple key genes, which ultimately determine the morphology, physiology, and functional identity of mDA neurons. This function of LMX1a is dependent on its homeobox domain. Here, we determined the structures of the LMX1a homeobox domain in complex with the promoter sequences of the Wnt family member 1 (WNT1) or paired like homeodomain 3 (Pitx3) gene, respectively. The complex structures revealed that the LMX1a homeobox domain employed its α3 helix and an N-terminal loop to achieve specific target recognition. The N-terminal loop (loop1) interacted with the minor groove of the double-stranded DNA (dsDNA), whereas the third α-helix (α3) was tightly packed into the major groove of the dsDNA. Structure-based mutations in the α3 helix of the homeobox domain significantly reduced the binding affinity of LMX1a to dsDNA. Moreover, we identified a nonsyndromic hearing loss (NSHL)-related mutation, R199, which yielded a more flexible loop and disturbed the recognition in the minor groove of dsDNA, consistent with the molecular dynamics (MD) simulations. Furthermore, overexpression of Lmx1a promoted the differentiation of SH-SY5Y cells and upregulated the transcription of WNT1 and PITX3 genes. Hence, our work provides a detailed elucidation of the specific recognition between the LMX1a homeobox domain and its specific dsDNA targets, which represents valuable information for future investigations of the functional pathways that are controlled by LMX1a during mDA neuron development.


Subject(s)
LIM-Homeodomain Proteins , Promoter Regions, Genetic , Transcription Factors , Wnt1 Protein , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/chemistry , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Protein Binding , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/chemistry , DNA/metabolism , DNA/genetics , DNA/chemistry , Protein Domains , Models, Molecular , Mutation , Crystallography, X-Ray , Binding Sites , Nucleotide Motifs
SELECTION OF CITATIONS
SEARCH DETAIL
...